US20130164216A1 - Bladder cancer tumor marker, antibody and use thereof - Google Patents

Bladder cancer tumor marker, antibody and use thereof Download PDF

Info

Publication number
US20130164216A1
US20130164216A1 US13/814,446 US201113814446A US2013164216A1 US 20130164216 A1 US20130164216 A1 US 20130164216A1 US 201113814446 A US201113814446 A US 201113814446A US 2013164216 A1 US2013164216 A1 US 2013164216A1
Authority
US
United States
Prior art keywords
bladder cancer
cells
bcmab1
antibody
integrin
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US13/814,446
Other versions
US9073995B2 (en
Inventor
Chong Li
Zusen Fan
Honglian Zhang
Zhonghua Dai
Haidong Tang
Jun Chen
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Institute of Biophysics of CAS
Original Assignee
Institute of Biophysics of CAS
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Institute of Biophysics of CAS filed Critical Institute of Biophysics of CAS
Assigned to INSTITUTE OF BIOPHYSICS, CHINESE ACADEMY OF SCIENCES reassignment INSTITUTE OF BIOPHYSICS, CHINESE ACADEMY OF SCIENCES ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHEN, JUN, DAI, ZHONGHUA, FAN, ZUSEN, LI, CHONG, TANG, HAIDONG, ZHANG, HONGLIAN
Publication of US20130164216A1 publication Critical patent/US20130164216A1/en
Application granted granted Critical
Publication of US9073995B2 publication Critical patent/US9073995B2/en
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2839Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the integrin superfamily
    • C07K16/2842Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the integrin superfamily against integrin beta1-subunit-containing molecules, e.g. CD29, CD49
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • A61K47/48607
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6861Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from kidney or bladder cancer cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1093Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody conjugates with carriers being antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70546Integrin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70546Integrin superfamily
    • C07K14/7055Integrin beta1-subunit-containing molecules, e.g. CD29, CD49
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3038Kidney, bladder
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/1048Glycosyltransferases (2.4)
    • C12N9/1051Hexosyltransferases (2.4.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/02Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
    • C12Q1/025Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/48Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving transferase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y204/00Glycosyltransferases (2.4)
    • C12Y204/01Hexosyltransferases (2.4.1)
    • C12Y204/01041Polypeptide N-acetylgalactosaminyltransferase (2.4.1.41)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57488Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving compounds identifable in body fluids
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6854Immunoglobulins
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70546Integrin superfamily, e.g. VLAs, leuCAM, GPIIb/GPIIIa, LPAM
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2440/00Post-translational modifications [PTMs] in chemical analysis of biological material
    • G01N2440/38Post-translational modifications [PTMs] in chemical analysis of biological material addition of carbohydrates, e.g. glycosylation, glycation

Definitions

  • the present invention belongs to a field of tumor immunology.
  • the present invention relates to a novel antigen AG- ⁇ 3 ⁇ 1 and the preparation thereof (AG: Aberrant Glycosylation), and an anti-AG- ⁇ 3 ⁇ 1 monoclonal antibody BCMab1. It is demonstrated at cytology and histology levels that AG- ⁇ 3 ⁇ 1 is expressed on the membranes of human bladder tumor cells only, the antibody BCMab1 not only specifically recognizes AG- ⁇ 3 ⁇ 1, but also effectively inhibits the proliferation of human bladder tumor cells at cellular level in vitro and in an animal model in vivo.
  • the present invention also relates to a process for detecting bladder cancer in human using a competitive ELISA.
  • the antigen in an immobile phase is human bladder cancer tumor marker AG- ⁇ 3 ⁇ 1 according to the present invention
  • the detection antibody is the antibody BCMab1 directed against human bladder cancer integrin ⁇ 3 ⁇ 1.
  • Bladder cancer is a tumor most commonly found in urinary systems. Although numerous steps were adopted for the treatment, 40-70% of the patients relapsed once or more than once and 10-15% of the patients developed more severe tumors or involved metastasis. Moreover, all these treating processes have apparent toxic and side-effects.
  • the medicaments used for preventing relapse by bladder perfusion are mainly divided into the following several classes: (1) anti-tumor chemotherapeutic agents, such as Mitomycin C, etc.; (2) immunopotentiators, such as Bacillus Calmette-Guerin Vaccine (BCG), etc.; and (3) cytokines, such as interferon, etc.
  • anti-tumor chemotherapeutic agents such as Mitomycin C, etc.
  • immunopotentiators such as Bacillus Calmette-Guerin Vaccine (BCG), etc.
  • BCG Bacillus Calmette-Guerin Vaccine
  • cytokines such as interferon
  • the above classes of medicaments not only can act on the entire bladder and urethra unspecifically, but also can be absorbed by the mucous membranes of bladder and urethra, which easily results in topical or systematic toxic and side-effects.
  • Mitomycin C and Bacillus Calmette-Guerin Vaccine which have more positive therapeutical effects
  • 90% of the patients who had received Bacillus Calmette-Guerin Vaccine were suffered from BCG cystitis and other side effects including hematuria, tetter, fever, arthritis, urethral stricture and the like, or infrequently, even some severe conditions, such as hepatitis, pneumonia, life-threatened septicemia and so on.
  • Mitomycin C has fewer side effects, however still 5-25% of the patients who had received Mitomycin C were suffered from chemical cystitis and anaphylaxis, and other side effects included urethral stricture, marrow depression, calcification of bladder wall, and so on.
  • Integrin is a group of divalent cation-dependent cell surface receptors with a major function of mediating cell-matrix, cell-cell adhesion by binding to corresponding ligands, so as to further affect cell shapes, gene expression and regulation, cell proliferation and differentiation, apoptosis, and migration, infiltration and metastasis of tumor cells, etc. Integrin is composed of non-covalent subunits ⁇ and ⁇ . Various combinations result in different ligand-binding abilities. Integrin subunit ⁇ 3 (CD49c) can be combined with subunit ⁇ 1 (CD29) to form integrin ⁇ 3 ⁇ 1.
  • the ligands for integrin ⁇ 3 ⁇ 1 are laminin (LN) and Collagen IV.
  • Integrin ⁇ 3 ⁇ 1 is mainly involved in the conglutination between cells and basilar membranes of epithelial cells and can adjust signal transduction, thereby impacting the biological features of cells. Integrin ⁇ 3 ⁇ 1 is modified aberrantly on the surface of tumor cells, which is considered to cause a change in malignancy.
  • Anti-tumor targeted drugs are biological or chemical molecules (e.g. monoclonal antibodies) with an ability to recognize and kill tumor cells specifically.
  • Bladder cancer is a tumor inside the body cavity of human, i.e. a “somatic test tube” of human body.
  • Targeted drugs have high specificities with and strong lethal effects on target cells in vitro. Therefore, in terms of the treatment for bladder cancer, it is very important to find a novel targeted drug directed against bladder cancer in human.
  • the inventors utilized human bladder cancer cell line T24 to immunize mice, so as to achieve hybridoma cells.
  • ELISA was conducted to screen for antibody BCMab1 which can bind to T24 cells with a high specificity. It was demonstrated by immunohistochemistry and immunofluorescence that antibody BCMab1 showed a strong positive reactivity with bladder cancer cell line T24 and bladder cancer tissue of human, but no cross reactivity with normal bladder tissue or other non-bladder cancer cells.
  • the antigen recognized by antibody BCMab1 was captured by antibody BCMab1 using an immunoaffinity chromatography, and then identified as integrin ⁇ 3 ⁇ 1 modified by aberrant glycosylation using a mass spectroscopy and carbohydrate chips.
  • Such an antigen is designated as AG- ⁇ 3 ⁇ 1, having an epitope in a carbohydrate structure shown as follow: [3OSO3]Gal ⁇ 1-4(Fuc ⁇ 1-3)[6OSO3]GlcNAc. It has been demonstrated by the antibody BCMab1 that AG- ⁇ 3 ⁇ 1 is only expressed on bladder tumor cells of human, and correlated positively with tumor staging and pathological grading of bladder cancer. AG- ⁇ 3 ⁇ 1 can be used as a novel marker for bladder cancer tumors of human.
  • T24 cells cultured in vitro and a nude mouse transplantation tumor model are treated with antibody BCMab1. It is found that antibody BCMab1 can effectively inhibit the proliferation of bladder cancer cell line T24. Antibody BCMab1 thus can be used as the targeted drug directed against bladder cancer of human.
  • a diagnostic reagent is made from antibody BCMab1 and antigen AG- ⁇ 3 ⁇ 1.
  • human bladder cancer tumor marker AG- ⁇ 3 ⁇ 1 as an immobile phase, competes with a sample to be tested for antibody AbBC1, so as to detect bladder tumor cells in human urine.
  • glycosyltransferase GALNT1 is highly expressed in bladder cancer and GALNT1-knockdown cells can down-regulate glycosylation level of integrin ⁇ 3 ⁇ 1.
  • Aberrant glycosylation of integrin ⁇ 3 facilitates malignant transformation of bladder cancer.
  • BCMab1 and immunotoxin BCMab1-Ra can significantly inhibit growth of the bladder cancer transplanted subcutaneously and orthotopically.
  • BCMab1 treatment can recruit and activate natural killer (NK) cells surrounding bladder cancer.
  • NK natural killer
  • Some creative ideas are brought up in the invention, including (1) preparing a mouse monoclonal antibody BCMab1 directed against human bladder cancer, which not only specifically binds to bladder cancer tissues of human, but also effectively inhibits proliferation of bladder cancer cell line T24 in vivo or in vitro; (2) revealing that the antigenic epitope AG- ⁇ 3 ⁇ 1 recognized by such an antibody is a novel marker for human bladder cancer tumors, and demonstrating that such an epitope is only expressed on bladder tumor cells of human and correlated positively with tumor staging and pathological grading of bladder cancer; and (3) developing a highly sensitive competitive ELISA method for detecting human bladder cancer.
  • the invention provides an aberrantly glycosylated integrin AG- ⁇ 3 ⁇ 1, as well as a hybridoma cell generating a monoclonal antibody directed against such a tumor marker and the monoclonal antibody BCMab1 secreted by the same.
  • the antigenic epitope recognized by the antibody BCMab1 is [3OSO3]Gal ⁇ 1-4(Fuc ⁇ 1-3)[6OSO3]GlcNAc.
  • Such a monoclonal antibody shows a strong positive reactivity with human bladder cancer cell line T24 and human bladder cancer tissues, but no cross reactivity with normal bladder tissue or other non-bladder cancer cells of human.
  • the monoclonal antibody has a function of inhibiting proliferation of bladder cancer cell line T24 in cell cultures in vitro and in animal tumor models.
  • the invention also provides an in vitro diagnostic kit comprising the monoclonal antibody BCMab1, and a process for detecting the content of the tumor marker in urine exfoliative cells using the monoclonal antibody BCMab1.
  • one object of the invention is to provide an aberrantly glycosylated integrin AG- ⁇ 3 ⁇ 1 characterized in that the integrin ⁇ 3 ⁇ 1 has a carbohydrate structure: [3OSO3]Gal ⁇ 1-4(Fuc ⁇ 1-3)[6OSO3]GlcNAc as an antigenic epitope.
  • the carbohydrate structure [3OSO3]Gal ⁇ 1-4(Fuc ⁇ 1-3)[6OSO3]GlcNAc is represented by a structural formula shown as follow:
  • the integrin ⁇ 3 ⁇ 1 has an ⁇ 3 subunit having an amino acid sequence shown as SEQ ID No: 1 and a ⁇ 1 subunit having an amino acid sequence shown as SEQ ID No: 2.
  • the aberrant glycosylation is located on the amino acid T (threonine) at position 740 of the ⁇ 3 subunit.
  • Another object of the invention is to provide a binding molecule directed against the AG- ⁇ 3 ⁇ 1 according to the invention.
  • a binding molecule can specifically recognize or bind to the carbohydrate structure [3OSO3]Gal ⁇ 1-4(Fuc ⁇ 1-3)[6OSO3]GlcNAc as the antigenic epitope.
  • the binding molecule is a polyclonal antibody or monoclonal antibody. More preferably, the binding molecule is a monoclonal antibody.
  • the binding molecule is an anti-AG- ⁇ 3 ⁇ 1 monoclonal antibody BCMab1, and the monoclonal antibody is secreted by the hybridoma cell line deposited as CGMCC No. 3845.
  • Another object of the invention is to provide a conjugate, wherein the binding molecule of the invention is conjugated with a substance selected from the group consisting of a biological marker, an antitumor drug, a toxin and a radioactive agent.
  • the conjugate is an immunotoxin BCMab1-Ra prepared from BCMab1 and ricin A chain connected by a heterogeneous difunctional crosslinker such as SPDP (N-succinimidyl 3-(2-pyridyldithio) propionate), MBS (m-Maleimidobenzoyl-N-hydroxysuccinimide ester), or 2-IT (2-Iminothiolane), etc.
  • SPDP N-succinimidyl 3-(2-pyridyldithio) propionate
  • MBS m-Maleimidobenzoyl-N-hydroxysuccinimide ester
  • 2-IT 2-Iminothiolane
  • connection method between an antibody and ricin A chain is well known and can be found in the following references for details: Shen, B. Connection between monoclonal antibody and toxin, Monoclonal Antibody Communication 1989; 2: 51; Cumber A J, et al. Preparation of antibodytoxin conjugates, Methods Enzymol 1985; 112:207; and Cros o, et al. Biochemical aspect of immunotoxin preparation, J. Immunol. Meth. 1985; 81:283.
  • Another object of the invention is to provide a pharmaceutical composition which comprises an effective dose of the binding molecule according to claims 3 or 4 or the conjugate according to claim 5 , and a pharmaceutically acceptable carrier, diluents or excipient.
  • a further object of the invention is to provide a kit for detecting bladder cancer, which comprises the binding molecule of the invention, the conjugate of the invention, or AG- ⁇ 3 ⁇ 1; and optionally another reagent for detecting bladder cancer.
  • the detection is carried out by an enzyme linked immunosorbent assay.
  • the enzyme linked immunosorbent assay is a competitive enzyme linked immunosorbent assay, wherein AG- ⁇ 3 ⁇ 1 of the invention, as an immobile phase, competes with a sample to be tested for the antibody of the invention.
  • the sample to be tested in said kit is urine or bladder tissue.
  • Another object of the invention is to provide a hybridoma cell line secreting the antibody BCMab1 directed against integrin ⁇ 3 ⁇ 1 of human bladder cancer, which is deposited as CGMCC No. 3845.
  • a further object of the invention is to provide use of the pharmaceutical composition of the invention in preparation of the medicament for the treatment of bladder cancer.
  • the medicament is used in a combination therapy with other drugs for the treatment of bladder cancer.
  • Said other drugs for the treatment of bladder cancer are preferably selected from the group consisting of antitumor chemotherapeutic agent, immunoreinforcing agent, and cytokines; and more preferably a group consisting of thiotepa (TSPAP), Mitomycin C (MMC), Bacillus Calmette-Guerin Vaccine (BCG), cisplatin (DDP), adriamycin (ADM) and interferon.
  • said medicament and said other drugs for the treatment of bladder cancer are used simultaneously, separately or sequentially for the treatment of human or animal bodies. Still preferably, said medicament and said other drugs for the treatment of bladder cancer, formulated in a same or different preparations, are co-administered to the subjects in need thereof through a same or different routes.
  • Another object of the invention is to provide use of the AG- ⁇ 3 ⁇ 1 or antibody of the invention, optionally combined with other indicators for the diagnosis or prognosis of bladder cancer, in the diagnosis and/or prognosis of bladder cancer.
  • Another object of the invention is to provide use of the AG- ⁇ 3 ⁇ 1 of the invention for preparing and screening antibodies, wherein the AG- ⁇ 3 ⁇ 1 is used as an immunogen to immunize animals, so as to obtain monoclonal or polyclonal antibodies, or the AG- ⁇ 3 ⁇ 1 is used as an antigen to screen and prepare antibodies by a phage, yeast or ribosome display technique.
  • Another object of the invention is to provide use of an RNAi sequence directed against GAL3ST2 in preparation of the medicament for the treatment of bladder cancer.
  • the nucleotide sequence of the RNAi is shown as SEQ ID No: 3.
  • Another object of the invention is to provide use of an inhibitor of N-acetylgalactosaminyltransferase 1 in preparation of the medicament for the treatment of bladder cancer.
  • the inhibitor includes the nucleotide sequence of the RNAi as shown by SEQ ID No: 4, uridine 5′-phosphoric (1-hexadecanesulfonic) anhydride, and Ammonium (3-Acetamido-2,6-anhydro-3-deoxy-D-glycero-L-gluco-heptitol-1-yl) (Uridine-5′-yl)Diphosphate.
  • Another object of the invention is to provide a process of screening for a potential substance to treat bladder cancer, which comprises the steps of:
  • Yet another object of the invention is to provide use of N-acetylgalactosaminyltransferase 1 in the diagnosis and/or prognosis of bladder cancer.
  • FIG. 1 BCMab1 specifically recognized bladder cancer cell line and bladder cancer tissue.
  • (a, b) It was shown by immunofluorescence and immunohistochemisty that BCMab1 stain was positive for bladder cancer cell line T24 cells and negative for normal bladder cancer cell line HCV29.
  • (c) BCMab1 stain was positive for bladder cancer tissues and negative for normal bladder mucosal tissues (IHC);
  • FIG. 2 Periodide-Oxidation Experiment (Experimental principle of the Periodic acid-Oxidation experiment: a hydroxyl group of the carbohydrate in glycoprotein is oxidized by a periodic acid in an acidic condition without any alteration of the polypeptide chain structure).
  • bladder cancer cells T24 were treated with iodic acid to destroy the carbohydrate antigen structure, so that antibody BCMab1 cannot recognize and no positive reaction with a brown color was present.
  • Anti-MUC1 antibody (commercially purchased from Novocastra Laboratories Ltd, UK) is an antibody known to bind to carbohydrate structures only, which was used as a control herein.
  • Anti-CK20 antibody (Novocastra Laboratories Ltd, UK) is an antibody known to bind to proteinic polypeptides only, which was also used as a control herein;
  • FIG. 3 Periodide-Oxidation Experiment (Dot blot hybridization experiment, with a result similar with FIG. 2 ). After lysis, bladder cancer cells T24 were spotted on a film and treated with iodic acid, so that the carbohydrate antigen structure was destroyed, carbohydrate antigen structure was not able to be detected by the antibody BCMab1 and no positive reaction was present;
  • FIG. 4 Results of the carbohydrate chip assay
  • FIG. 5 Knockdown of integrin ⁇ 3 or integrin ⁇ 1 in T24 cells (Vector is a vector control, and ⁇ -actin is an internal standard for amplification);
  • FIG. 6 Integrin ⁇ 3-knockdown T24 cells were barely stained by BCMab1, whereas integrin ⁇ 1-knockdown T24 cells were stained (shIntegrin ⁇ 3 and shIntegrin ⁇ 1 represent the cells in which the ⁇ 3 and ⁇ 1 subunits are knocked down by RNAi, respectively);
  • FIG. 7 Impact of GAL3ST2 on aberrant glycosylation of integrin ⁇ 3 ⁇ 1 in bladder cancer tissues.
  • Knockdown of GAL3ST2 could reduce the colony forming ability of T24 cells significantly.
  • (e) Knockdown of GAL3ST2 could reduce the adhesion of T24 cells significantly.
  • FIG. 8 Quantitative RT-PCR detection for GALNT1 silence effect in EJ cells
  • FIG. 9 GALNT1 silence almost blocked BCMab1 staining.
  • T24 cells and an inhibitor for GALNT1, i.e. BG were co-incubated for 7 days, and then stained with BCMab1;
  • FIG. 10 Knockdown of integrin ⁇ 3 or GALNT1 inhibited cancer cell proliferation and tumor growth.
  • Integrin ⁇ 3- or GALNT1-silenced cell lines could not grow in a soft agar. After a treatment with BCMab1 (10 ⁇ g/ml), the growth of T24 cells was also inhibited in a soft agar.
  • FIG. 11 Knockdown of integrin ⁇ 3 inhibited the adhesion between tumor and extracellular matrix (ECM) and tumor migration, as well as facilitated NK cell-mediated cell killing.
  • ECM extracellular matrix
  • FIG. 11 Knockdown of integrin ⁇ 3 inhibited the adhesion between tumor and extracellular matrix (ECM) and tumor migration, as well as facilitated NK cell-mediated cell killing.
  • a, b Compared with an empty vector, knockdown of integrin ⁇ 3 weakened the adhesion between tumors and ECM (a) and tumor migration (b) (p ⁇ 0.01); whereas knockdown of GALNT1 failed to show an evident result.
  • BCMab1 (10 ⁇ g/ml) could inhibit cell junction and tumor migration significantly (mIgG as negative control).
  • Tumor volumes were measured periodically, and averaged (the left panel). The tumor status on Day 35 is shown in the right panel.
  • FIG. 12 Antibody BCMab1 inhibited the proliferation of tumor cells
  • FIG. 13 Amino acid sequence of integrin ⁇ 3 subunit
  • FIG. 14 Amino acid sequence of integrin ⁇ 1 subunit
  • FIG. 15 BCMab1 and BCMab1-Ra inhibited growth of the transplanted tumors of a mouse-borne tumor model.
  • BCMab1 and BCMab1-Ra inhibited cancer growth.
  • T24 cells were injected into nude mice subcutaneously. When the tumor diameter reached 3-5 mm, the mice were grouped (10 mice/group), and injected with BCMab1 or BCMab1-Ra (10 mg/kg) intraperitoneally, 3 times/week, for 5 weeks. The tumor statue on Day 35 is shown in the lower panel.
  • FIG. 16 BCMab1 resulted in cell cycle arrest and inhibited tumor growth.
  • BCMab1 resulted in cell cycle arrest.
  • BCMab1 (10 ⁇ g/ml)-treated T24 cells were analyzed by a PI (Propidium Iodide)-staining flow cytometer. mIgG was used as a negative control.
  • PI Propidium Iodide
  • mIgG was used as a negative control.
  • Antibody BCMab1 inhibited phosphorylation of FAK and c-Jun, and downregulated expression of cyclin D1 and CDK4.
  • the lysate was immunoblotted by anit-phospho-FAK (Tyr397) and anti-phospho-c-Jun (Ser63 and Ser73) as well as the total FAK and c-Jun. At the same time, the expression of cyclin D1 and CDK4 was detected. ⁇ -actin was used a control.
  • T24 cells and integrin ⁇ 3- or GALNT1-silenced T24 cells were treated with Matrigel in a serum-free condition for 30 min. HCV29 cells were used as a normal control.
  • BCMab1 still could inhibit tumor growth.
  • mice After removal of NK cells using anti-asialo GM1 (GM1) antibody, the mice were administered with BCMab1, or inoculated with Integin ⁇ 3-knockdown T24 cells. On Day 35, tumor volumes were measured, and averaged (the left panel). The right panel shows the tumor tissues;
  • FIG. 17 Aberrant glycosylation of integrin ⁇ 3 ⁇ 1 was correlated with clinical severity and prognosis.
  • the expression level of integrin ⁇ 3 ⁇ 1 was high in the bladder cancer specimen with a higher grade, deeper invasion and lymph-node involvement; whereas low in the case with a lower grade, superficial invasion and lymph-node involvement-free.
  • binding molecules means intact immunoglobulins, including monoclonal antibodies and polyclonal antibodies, such as chimeric antibodies, humanized antibodies or human monoclonal antibodies, or antigen binding fragments which can bind to antigenic epitopes, including Fab, F(ab′), F(ab′)2, Fv, dAb, Fd, complementary determining region (CDR) fragment, single-chain antibodies (scFv), bivalent single-chain antibodies, single-chain phage antibodies, bispecific double-chain antibodies (diabodies), triabodies, tetrabodies, and proteins or polypeptides binding to antigenic epitopes, etc.
  • monoclonal antibodies and polyclonal antibodies such as chimeric antibodies, humanized antibodies or human monoclonal antibodies, or antigen binding fragments which can bind to antigenic epitopes, including Fab, F(ab′), F(ab′)2, Fv, dAb, Fd, complementary determining region (CDR) fragment, single-chain antibodies (s
  • the above fragments can be generated through synthesis, enzymatic or chemical lysis of intact immunoglobulins, or recombinant DNA technology-genetic engineering. All these preparation methods are well known in the art, for example, E. Harlow and D, Lane (1988) ed. Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., which is incorporated herein by reference.
  • binding molecules include all classes and subclasses of immunoglobulins known in the art. According to the amino acid sequences of their heavy-chain constant domains, binding molecules can be divided into 5 major classes of intact antibodies: IgA, IgD, IgE, IgG and IgM, wherein some classes can be further divided into subclasses (isotypes), e.g. IgA1, IgA2, IgG1, IgG2, IgG3 and IgG4.
  • An anti-AG- ⁇ 3 ⁇ 1 monoclonal antibody, BCMab1 is provided in the invention, and anti-AG- ⁇ 3 ⁇ 1 polyclonal antibodies and other monoclonal antibodies can be easily generated by a person skilled in the art.
  • the general processes for preparing a monoclonal antibody from hybridoma are well known.
  • the cell lines that generate immortal antibodies can be prepared through cell fusion and other techniques, such as transformation of B lymphocytes by oncogenic DNA, or EB (Epstein-Barr virus) transfection.
  • Physical selection is one example of the conventional processes for obtaining all human antibodies, and includes but is not limited to screening by a phage, yeast or ribosome display technique.
  • the detailed procedure comprises a step of selecting a ligand (antibody) binding to the target protein (antigen) accordingly.
  • ligand antibody
  • phage, bacterium and yeast display techniques a living organism is used for the amplification and display, and phenotype and genotype are combined; whereas ribosome and puromycin mRNA display techniques rely on PCR amplification, in vitro RNA translation for obtaining a ligand, and then the binding to corresponding cDNA via a noncovalent bond or a covalent bond. These methods usually require several rounds of screenings. Since the antigenic specificities are known in advance, therefore the specificities of the resultant antibodies are definite. All these methods are well known by a person skilled in the art, and described detailedly in Antibody Engineering Drugs (Zhen, Y. and Shao, R., Chemical Industry Press, published on 2002 Nov. 1).
  • the invention first reveals a relation between the increase of AG- ⁇ 3 ⁇ 1 and development/prognosis of bladder cancer, and demonstrates that AG- ⁇ 3 ⁇ 1 can be used for the diagnosis, therapeutic direction and prognosis. It is understood by a person skilled in the art that all kinds of methods, including but not limited to immunohistochemistry, immunoenzyme technique, enzyme linked immunosorbent assay, immunofluorescence technique, radioimmunoassay, immune-colloidal gold labeling technique and so on, can be applied to detect AG- ⁇ 3 ⁇ 1.
  • markers which can be detected commonly by binding to the detection antibodies in the field.
  • the markers applied in the invention are not specifically limited, as long as they can bind to the detection antibodies and, after treated properly, accurately indicates whether AG- ⁇ 3 ⁇ 1 is present in the samples to be detected and the existing amount thereof.
  • the markers can be selected from (but not limited to): horseradish peroxidase, alkaline phosphatase, glucose oxidase, ⁇ -D-galactosidase, urease, catalase, or glucoamylase.
  • the substances include (but are not limited to): o-phenylene diamine (OPD), tetramethyl benzidine (TMB) and ABTS for horseradish peroxidase; and p-nitrophenyl phosphate (p-NPP) for alkaline phosphatase.
  • OPD o-phenylene diamine
  • TMB tetramethyl benzidine
  • ABTS horseradish peroxidase
  • p-NPP p-nitrophenyl phosphate
  • a quality control i.e. control
  • the quality control is prepared by diluting a standard AG- ⁇ 3 ⁇ 1 preparation and serum obtained from healthy volunteers.
  • a person skilled in the art understands that several standard AG- ⁇ 3 ⁇ 1 preparations with known concentrations can be set up during the detection.
  • the standard preparations can be set up by conventional methods.
  • sample means a substance extracted from human or animal bodies, and includes but is not limited to: urine, plasma, serum, bladder tissue, and other tissues or extraction liquids thereof.
  • sample is urine.
  • bladder cancer cells release various types of soluble molecules, including AG- ⁇ 3 ⁇ 1.
  • the invention further provides a pharmaceutical composition, which comprises:
  • composition of the invention includes pharmaceutical compositions, as long as they comprise the binding molecules of the invention as an active component.
  • the term “comprise” means that all types of components can be applied to the mixture or composition of the invention.
  • a “pharmaceutically acceptable” component is a substance which is suitably used in human and/or animals without causing excessive adverse side effect (such as toxicity, irritation and allergy), i.e. a substance with a reasonable efficiency/risk ratio.
  • an effective amount means the amount of a therapeutical agent to treat, alleviate or prevent diseases or conditions, or the amount showing detectable therapeutical or prophylactic effect.
  • the accurate effective amount for a particular subject depends on its somatotype and healthy condition, feature and severity of the disease, as well as the selected therapeutical agents and/or the combination thereof for administration. As a result, predetermination of the accurate effective amount is meaningless.
  • the effective amount can be determined by a clinical physician through conventional experiments.
  • the effective amount means administration of 0.01 ⁇ g/kg-500 ⁇ g/kg, preferably 0.05 ⁇ g/kg-200 ⁇ g/kg body weight of the active substance of the invention into individuals.
  • the pharmaceutical composition can further comprise a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier means a carrier used for administering a therapeutical agent.
  • a pharmaceutically acceptable carrier satisfies the following criteria: it neither induces generation of the antibodies which harm the individuals who have received the composition, nor causes excessive toxicity upon administration.
  • Such carriers are well known by a person skilled in the art. A sufficient discussion of the pharmaceutically acceptable carriers can be found in Remington's Pharmaceutical Sciences, Mack Pub. Co., N.J. 1991.
  • Solid carriers include but are not limited to: starch, lactose, dicalcium phosphate, microcrystalline cellulose, sucrose and kaolin; and liquid carriers include but are not limited to: saline, buffer, glucose, water, glycerol, ethanol, adjuvant, polyethylene glycol, nonionic surfactant and edible oil (such as corn oil, peanut oil and sesame oil), as long as they are suitable for the properties of the active components and the desired specific administration routes.
  • the adjuvants commonly used for preparing pharmaceutical compositions can also be included advantageously, for example, a flavoring agent, pigment, preservative and antioxidant such as vitamin E, vitamin C, BHT and BHA. Additionally, these carriers possibly further comprise auxiliaries such as a wetting agent or emulsifying agent, pH buffer substance and so on.
  • the pharmaceutical composition of the invention can be prepared into any conventional preparation forms by conventional methods.
  • the dosage forms of the pharmaceutical composition according to the invention are various, as long as they enable the active component to enter into mammalian bodies effectively.
  • the composition can be formulated into any forms suitable for the desired administration routes.
  • the pharmaceutical composition can be made into tablets, pills, powder, lozenges, pouches, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosol (as a solid or in a liquid matrix), ointment, soft and hard gel capsules, suppositories, aseptic injections, aseptic packagings of powder, and so on.
  • binding molecules or their pharmaceutically acceptable derivatives, modifications, and combinations thereof can be administered through bladder infusion, intravenous, intramuscular or subcutaneous injection, and an oral route, etc., preferably through bladder infusion.
  • the composition of the invention and the pharmaceutical composition thereof can also be stored in a disinfector suitable for injection or dripping.
  • composition of the invention once prepared, can be given to a subject directly.
  • the subjects to be prevented or treated can be animals; particularly human.
  • the therapeutical dose regimen can be a single-dose regimen or a multidose regimen, which can be adjusted for providing an optimal therapeutical response. For example, due to the stringent requirements of the therapeutical status, several separated doses can be given every day, or the dose can be reduced proportionally.
  • composition or medicament of the invention can further comprise other active components, e.g. other drugs for the treatment of bladder cancer used in a combination therapy.
  • Said other drugs for the treatment of bladder cancer are selected from preferably the group consisting of antitumor chemotherapeutic agent, immunoreinforcing agent, and cytokines; and more preferably a group consisting of thiotepa (TSPAP), Mitomycin C (MMC), Bacillus Calmette-Guerin Vaccine (BCG), cisplatin (DDP), adriamycin (ADM) and interferon.
  • TSPAP thiotepa
  • MMC Mitomycin C
  • BCG Bacillus Calmette-Guerin Vaccine
  • DDP cisplatin
  • ADM adriamycin
  • interferon where two or more drugs mentioned above are administered in a combination, normally an effect that excels those caused by administering the two drugs separately is achieved.
  • GALNT1 N-acetylgalactosaminyltransferase 1
  • the inhibitors include but are not limited to the nucleotide sequence (SEQ ID No: 4) of RNAi mentioned in Examples, an inhibitor for GALNT1, i.e.
  • GALNT1 mRNA expression level was higher in bladder cancer tumor tissues than in normal tissues. Therefore, GALNT1 mRNA can be used as indicator for diagnosis and/or prognosis of bladder cancer.
  • Conventional methods for detecting GALNT1 mRNA are known by a person skilled in the art. Specifically, the tissue to be detected and the normal control tissue are obtained, and detected for their GALNT1 mRNA contents; if the GALNT1 mRNA level is higher in the tissue to be detected than in the normal control tissue by 2 folds or more, then the tissue to be detected is indicated to be a bladder cancer tissue.
  • GAL3ST2 galactose: 3-O-sulfonyltransferase 2
  • the inhibitors include but are not limited to the nucleotide sequence (SEQ ID No: 3) of RNAi mentioned in Examples and an RNAi inhibitor for GAL3ST2, i.e. 5′atgtggttcgacttcggct3′ disclosed by Xing-Zhong Wu (Biochemical and Biophysical Research Communications 332 (2005) 934-940), etc.
  • mice (commercially purchased from Vital River Laboratory Animal Technology Co. Ltd., Beijing) were immunized with human bladder cancer cell line T24 (ATCC: HTB-4) by intraperitoneally injecting a dose of 1 ⁇ 10 7 cells/ml PBS. After two weeks, the mice were immunized again with the same injection volume and method. While the serum titer of the mice met the requirement, cell fusion was arranged. The mice were boosted three days before the fusion. At the same time, mouse myeloma cells Sp2/0 (ATCC CRL-1772) were prepared.
  • the sensitized B lymphocytes were fused with the myeloma cells (Practical Immunology, Yang, Y. ed., Changchun Press, published in December, 1994), and selectively cultured (using peritoneal macrophages of the mice as feeder cells) in HAT medium (purchased from Invitrogen Co.; HAT is an enumerate of the initials of hypoxantin, aminopterin and thymidin; HAT medium is thus a cell culture medium containing these three substances).
  • the supernatant of the hybridoma cell culture was detected by ELISA. More specifically, T24 cells (1 ⁇ 10 5 cells/ml) were coated onto a 96-well plate, 100 ⁇ l/well, and cultured at 37° C. overnight. After cell adherence, 4% paraformaldehyde was added, and the plate was fixed at room temperature for min, then washed for 3 times. Subsequently, 100 ⁇ l of the supernatant to be detected was added and incubated at 37° C. for 1 hour. Following 3 times of washing, an enzyme-labeled secondary antibody (anti-mouse IgG-HRP) (commercially purchased from Zhongshanjinqiao Biological technology Co.
  • anti-mouse IgG-HRP enzyme-labeled secondary antibody
  • the selected positive hybridoma cells were clonally cultured (through a limiting dilution, and using peritoneal macrophages of the mice as feeder cells). Following 2-3 rounds of clonal cultures, stable hybridoma cell clones that could generate high-titer monoclonal antibodies were obtained. The hybridoma cell clones were then amplified, cultured, frozen and stored.
  • hybridoma cell line secreting monoclonal antibodies directed against human bladder cancer.
  • This hybridoma cell line was deposited in China General Microbiological Culture Collection (CGMCC, Beijing, China), with an accession number CGMCC No. 3845, on May 21, 2010.
  • the above hybridoma cells which secrete BCMab1 were inoculated into the abdominal cavities of Balb/C mice, so as to prepare ascites; then the monoclonal antibodies were extracted from the ascites.
  • the monoclonal antibody BCMab1 was purified using Protein G affinity chromatography. First, a Protein G affinity chromatographic column (purchased from “GE” Co.) was prepared, and then balanced with PBS (Phosphate Buffered Saline). The ascites containing monoclonal antibody BCMab1 was passed through the column, then the column was washed with PBS till its OD value approached zero.
  • the column was then eluted with a 0.2 M glycine-HCl solution (pH 2.8), and the eluent was collected. After the OD value of each collecting tube was measured, the eluent with the OD value within the peak range was retained, concentrated by dialysis, then frozen and stored at 20° C.
  • Normal bladder tissue cells HCV29 were purchased from Peking University Health Science Center. Cell lines T24, EJ, HepG2, LoVo, 293, A375, Jurkat, PC-1, MCF-7, K562, Hela, and other cell lines were purchased from ATCC (Rockville, Md., US). Bladder cancer tissues and normal tissues of human were received from The Second affiliated Hospital of Kunming Medical University, with informed consents. BALB/C mice and nude mice were both purchased from Chinese Academy of Medical Science Institute of laboratory animal.
  • Bladder cancer tissue slices of human were stained immunohistochemically with the monoclonal antibody BCMab1 prepared in Example 1 using a conventional method.
  • the result, as shown in FIG. 1 indicated that the bladder cancer tissues of human showed positive reactivity after stained with the monoclonal antibody BCMab1, a secondary antibody (anti-mouse IgG-HRP) and DAB substrate (Zhongshanjinqiao Biological technology Co. Ltd., Beijing); whereas the normal tissues of human showed negative reactivity after being stained with the monoclonal antibody BCMab1, a secondary antibody and substrate.
  • Bladder cancer cells T24 and other cells were detected by the monoclonal antibody BCMab1 using a flow cytometer, and normal human tissues were assayed immunohistochemically.
  • the results, as shown in Table 1 and FIG. 1 indicated that the monoclonal antibody BCMab1 showed a strong positive reactivity with T24 and EJ cells (ATCC CRL-1888), but no cross reactivity with other non-bladder cancer cells or normal tissues of human.
  • BCMab was able to stain human bladder cancer cell lines EJ and T24, rather than nonmalignant uretal epithelial cell line HCV29 and systematic tumor cells ( FIG. 1 a , 1 b and Table 1).
  • BCMab1 In order to investigate the specificity of BCMab1 to bladder cancer, 123 tumor tissues and 56 normal tissues were screened immunohistochemically. The results indicated that BCMab1 stained bladder cancer tissues only, and showed negative staining for normal bladder tissues and other tissues, such as liver, brain, adrenal gland, pancreas, stomach, colon, breast, lung, ovary, cardiac muscle, thyroid gland, lymph node and marrow ( FIG. 1 c , Table 1, unshown data). As a result, BCMab1 can specifically recognize the antigen on membranes of bladder cancer cells.
  • Human bladder cancer cell line T24 was cultured in a RPMI-1640 medium containing 10% fetal bovine serum (purchased from Invitrogen Co.). After digested with 0.25% pancreatin, 1 ⁇ 10 8 T24 cells were washed with PBS for 3 times. Then 100 ⁇ g monoclonal antibody BCMab1 was added, incubated at 4° C. for 2 hours, and washed with PBS for 3 times.
  • the cells were lysed with 1 ml detergent lysis buffer (50 mM Tris-HCl, pH 8.0; 150 mM NaCl; 0.02% sodium azide; 0.1% SDS; 100 ⁇ g/ml PMSF; 1 ⁇ g/ml Aprotinin; 1% NP-40; 0.5% sodium deoxycholate) for 30 min, and centrifuged at 12,000 g for 10 min. The supernatant was separated and passed through the Protein G affinity chromatographic column. The column was washed with PBS till its OD value approached zero. Subsequently, elution was carried out using a 0.2 M glycine-HCl solution (pH 2.8), and the eluent was collected. The OD value of each collecting tube was measured, and then the eluent with an OD value within the peak range was retained, and identified to be human integrin ⁇ 3 ⁇ 1 by mass chromatographic analysis (see Table 2).
  • 1 ml detergent lysis buffer 50 mM Tris
  • Mass spectrometry protein gel was cut into small pieces. Pierce Silver Staining Kit and mass chromatographic analysis (Pierce, US) were utilized. Mass chromatographic analysis was conducted for the tryptic peptide generated from digestion of the gel (NanoLC-LTQ, Thermo Finnigan).
  • T24 cells were cultured in a 24-well plate for 24 hours. A pre-cooled paraformaldehyde solution (1 ml) was added and kept for 15 min to fix the cells. T24 cells were oxidized by the high-iodine protein in a sodium-acetate balanced solution at room temperature for 1 hour. Excessive periodate was neutralized after cultured with sodium borohydride at room temperature for 30 min. T24 cells were stained immunohistochemically with antibodies BCMab1, anti-MUC1 or anti-CK20. The secondary antibody labeled with peroxidase and DAB assay kit were used for color development.
  • BCMab1 was diluted with 3% BSA/PBS buffer to a concentration of 1 ⁇ g/ml.
  • 100 ⁇ l BCMab1 diluention was added and incubated in a wet box for 1 hour.
  • the glass slide was rinsed with 0.05% Tween 20/PBS (PBS-T), and incubated with 200 ⁇ l Cy5-linked goat anti-mouse IgG antibody (1:200) (Novocastra Laboratories Ltd, UK) in a wet box for 1 hour.
  • the slide was dried in air, scanned to detect fluorescence at a wavelength of 635 nm using a microarray scanner (GenePix 4000B, Molecular Devices), and analyzed with the GenePix Pro software.
  • the experiment for determining the specific position of the polysaccharide epitope recognized by BCMab1 in integrin ⁇ 3 ⁇ 1 subunits is shown as follows: ⁇ 3 or ⁇ 1 subunit was knocked down by RNAi (through pSUPER-shIntegrin ⁇ 3 or pSUPER-shIntegrin ⁇ 1 viral transfection). ⁇ 3 or ⁇ 1 subunit-silenced T24 cells and stable cell lines were screened by puromycin ( FIG. 5 ). ⁇ 3 or ⁇ 1 could be knocked down by 80%. The ⁇ 3 or ⁇ 1 subunit-silenced T24 cells were stained by antibody BCMab1 and analyzed by FACS.
  • FACS flow cytometry is shown as follow: first, a single-cell suspension was prepared, and stained on ice for 40 min by adding antibodies; the cells were washed with the PBS containing 0.1% BSA for 3 times, and incubated with FITC-labeled secondary antibody on ice for 40 min; after washing, an analysis was conducted using FACSCalibur flow analysis system (BD, US).
  • RNAi knocking down integrin ⁇ 3 ⁇ 1 by RNAi
  • a small interfering RNA system acting on integrin 3 or ⁇ 1 was established using pSUPER (Oligoengine, Washington, US) and then cloned into a pSUPER system, so as to express a 19 nt hairpin structure (shRNA) (comprising a small loop of 9 nt).
  • shRNA 19 nt hairpin structure
  • T24 cells were transfected by Lipofectamin2000 (Invitrogen, US). Integrin ⁇ 3 ⁇ 1- and GALNT1-knockdown cells were screened out using puromycin. The cells transfected with empty vectors were used as a control.
  • the antigen AG- ⁇ 3 ⁇ 1 according to the invention can also be prepared by the following standard protocol:
  • GAL3ST galactose: 3-O-sulfonyltransferase
  • GAL3ST including 4 members i.e. GAL3ST1, GAL3ST2, GAL3ST3 and GAL3ST4, could move sulphates to the position Gal C-3.
  • GAL3ST2 galactose: 3-O-sulfonyltransferase 2
  • FIG. 7 Knockdown of GAL3ST2 could significantly inhibit the growth and migration of cancer. All these results suggested that GAL3ST2 played an important role in the aberrant glycosylation of integrin ⁇ 3 ⁇ 1 in bladder cancer cells and carcinogenesis.
  • GAL3ST2 Knockdown of GAL3ST2 was conducted as follows. A small interfering RNA system acting on GAL3ST2 was established using pSUPER and then cloned into a pSUPER system, so as to express a 19 nt hairpin structure (shRNA) (comprising a small loop of 9 nt). The sequences encoding GAL3ST2 shRNA
  • the inserted shRNAs (pSUPER-shGAL3ST2) were confirmed in these DNA sequences.
  • T24 cells were transfected by Lipofectamin2000 (Invitrogen, US).
  • GAL3ST2-knockdown cells were screened out using puromycin.
  • the cells transfected with empty vectors were used as a control.
  • 5′-GATCCCC TCCGAAACCTGCTCTTCTT TTCAAGAGA AAGAAGA GCAGGTTTCGGA TTTTTA-3′ (GAL3ST1, sense); 5′-AGCTTAAAAA TCCGAAACCTGCTCTTCTT TCTCTTGAA AAGA AGAGCAGGTTTCGGA GGG-3′ (GAL3ST1, antisence).
  • Matrigel (BD, US) was coated on a 96-well plate, and blocked with 10 mg/ml thermally inactivated BSA for 1 hour. After becoming confluence, the above knockdown T24 cells were digested with 1% trypsin, collected at a concentration of 5 ⁇ 10 5 cell/ml, resuspended in RPMI-1640 containing 1% FBS, and reconstituted at 37° C. for 15 min. shRNA vector was used as a control. The unadhered cells were rinsed off, whereas the adhered cells were collected and stained with 0.1% crystal violet (Sigma, Germany). After decoloration and dye solubilization, the samples were read at 575 nm by ELISA reader.
  • Transwell experiment utilized optimized Boyden chemotaxis kit (with an pore size of 8 ⁇ m, Costar, US). Matrigel (100 ⁇ g/ml) was coated, and blocked with the PBS containing 1% BSA. The above knockdown T24 cells were digested with 1% trypsin, washed, and resuspended in a medium containing 1% BSA. The lower chambers contained 10% FBS or 1% BSA (negative control). After being incubated at 37° C. for 4 hour, the cells remained in the upper chambers were sucked out, and the cells transferred into the lower chambers were fixed with ethanol and stained with Giemsa. The cells that penetrated through the filter membrane were counted under a microscope. Mitomycin C (2 ⁇ g/m) was used to inhibit cell proliferation.
  • GALNT1 N-Acetyl-Galactosaminyltransferase 1
  • the bladder cancer tissue specimens were labeled with Cy3; whereas the normal specimens were labeled with Cy5.
  • the labeled cDNA probes were mixed with 2.5 ⁇ SSC, 0.2% SDS, and 1 ⁇ g PolyA RNA.
  • the microarray chips were hybridized at 50° C. for 16 hours, and analyzed by ScanArray 4000 Microarray Analysis System (Packkad Bioscience, US). The data were analyzed by Microarray Analysis Software (Dapple version 0.68 beta).
  • GALNT1 in bladder cancer tissues Normal bladder tissues Bladder tumor tissues GenBank No. Gene name Signal ⁇ Detection ⁇ Signal ⁇ Detection ⁇ Fold-change # NM_020474 GALNT 1 303 ⁇ 26 P 3384 ⁇ 142 P 11.2 NM_004481 GALNT 2 748 ⁇ 23 P 703 ⁇ 35 P 0.9 NM_004482 GALNT 3 1854 ⁇ 72 P 1315 ⁇ 118 P 0.7 NM_003774 GALNT 4 36 ⁇ 3 A 42 ⁇ 7 A 1.2 NM_014568 GALNT 5 12 ⁇ 1 A 16 ⁇ 1 A 1.3 NM_007210 GALNT 6 815 ⁇ 39 P 902 ⁇ 45 P 1.1 NM_017423 GALNT 7 735 ⁇ 43 P 836 ⁇ 72 P 1.1 NM_017417 GALNT 8 87 ⁇ 9 A 107 ⁇ 23 A 1.2 NM_021808 GALNT 9 172 ⁇ 26
  • T24 cells were transfected with pSUPER-shGALNT1 vectors, so as to downregulate GALNT1 expression.
  • the GALNT1-knockdown T24 cell lines that had been transfected stably were screened out using puromycin ( FIG. 8 ).
  • GALNT could be silenced by 80%.
  • the GALNT1-knockdown cell lines could downregulate BCMab1 staining signal significantly ( FIG. 9 , left).
  • a competitive inhibition experiment was conducted to the substrate BG of GALNT1.
  • signal of integrin ⁇ 3 ⁇ l recognized by BCMab1 was downregulated ( FIG. 9 , right).
  • mIgG was the negative control. The result showed that GALNT1 might induce aberrant methylation of the integrin ⁇ 3 ⁇ 1 in bladder cancer.
  • GALNT1 The method for silencing GALNT1 is shown below.
  • a small interfering RNA system acting on GALNT1 was established using pSUPER and then cloned into a pSUPER system, so as to construct a pSUPER-shGALNT1 vector and express a 19 nt hairpin structure (shRNA) (comprising a small loop of 9 nt).
  • shRNA 19 nt hairpin structure
  • the inserted shRNAs (pSUPER-shGALNT1) were confirmed in these DNA sequences.
  • T24 cells were transfected by Lipofectamin2000 (Invitrogen, US). GALNT1-knockdown cells were screened out using puromycin. The cells transfected with empty vectors were used as a control.
  • GALNT1 was inhibited by BG.
  • T24 cells were cultured in a 6-well plate with or without 2 mM BG (Sigma, Germany) for 7 days. The cells were collected, into which BCMab1 and its corresponding FITC-linked secondary antibody were added and incubated for 40 min. The expression of antigen of BCMab1 was analyzed by FACS.
  • Knockdown of integrin ⁇ 3 or GALNT1 could significantly reduce the binding rate between [ 3 H]-thymidine and DNA of T24 cells ( FIG. 10 , a), and remarkably affect the cloning of T24 cells ( FIG. 10 , b).
  • a tumor-bearing model was constructed by inoculating BALB/c nude mice at their backs subcutaneously, then tumor change was observed by injecting vectors.
  • the integrin ⁇ 3- or GALNT1-knockdown group significantly inhibited growth of the subcutaneous tumors ( FIG. 10 , c). And by the end of the observation period, i.e. on Day 35, the tumor inhibition ratio reached 63% and 42%, respectively.
  • the steps of the cell proliferation experiment are shown as follows. 3 H nuclide incorporation assay was utilized. Wildtype T24 cells and shIntegrin ⁇ 3- or shGALNT1-knockdown T24 cells (2 ⁇ 10 3 cells/well) were coated onto a 96-well plate and cultured till reaching 70%-80% confluence. The cells were cultured in serum-free RPMI-1640 for 24 hours and the medium was replaced by RPMI-1640 containing 10% FBS. To the wildtype T24 cells, 10 ⁇ g/ml of BCMab1 or control mIgG was added. After 72 hours, 3 H was added and the cells were cultured for 4 hours. Then the cells were collected and detected for 3 H incorporation amount by a liquid scintillation counter LKB1219.
  • mice Female, 6 weeks old, with a body weight of about 15 g
  • T24 cells including wildtype T24 cells and shIntegrin ⁇ 3- or shGALNT1-knockdown T24 cells
  • tumor cells (1 ⁇ 10 7 , suspended in PBS) were injected subcutaneously at their backs.
  • tumors grew up to 3-5 mm
  • the mice with wildtype T24 were grouped (10 per group), and BCMab1 or mIgG (10 mg/kg) was injected intraperitoneally, 3 times/week, for 35 days.
  • the tumor volumes were measured twice per week.
  • Orthotopic bladder cancer transplantation model T24 cells were labeled with GFP, and tumors were induced to be transplanted into bladders by blood vessel probes.
  • BCMab1 or mIgG (1 mg/kg) was administered into the bladders of 24 mice in each group.
  • the images were obtained by an in vivo imaging system (IVIS) on days 5, 10, 15, 20, 25 and 30 after transplantation.
  • IVIS in vivo imaging system
  • NK cells were eliminated by anti-asialo GM1 antibody (Wako Chemicals, Richmond, Va.) in BALB/C mice before inoculation of T24 cells.
  • anti-asialo GM1 antibody Wi-Fi Protected Cells
  • integrin ⁇ 3- or GALNT1-knockdown T24 cells could significantly inhibit growth of tumors, compared with empty vectors.
  • No died cell was found in the tumor tissues of ⁇ 3- or GALNT1-T24-inoculated mice from which NK cells had been eliminated ( FIG. 11 , f); whereas a few died cells were observed in the early-stage tumor tissues from which NK cells had not been eliminated.
  • NK cells could significantly facilitate the expression of perforin and IFN ⁇ ( FIG. 11 , g, h). Moreover, the number of the NK cells was greater in shIntegrin ⁇ 3 or shGALNT1-treated tumors than in the control group ( FIG. 11 , i). The data suggested that the killing action mediated by NK cells played an important role in the tumor-bearing mouse with ⁇ 3- or GALNT1-knockdown T24.
  • T24 cells were inoculated into a 96-well plate in a density of 1 ⁇ 10 3 cells/well, and cultured conventionally till reaching the confluence of 70-80%. Then the cells were starved in a serum-free medium for 24 hours. Antibody BCMab1 (100 ⁇ g/mL) was added, and the culture was continued for 48 hours while serum was resumed. At the same time, homogenic mouse IgG (mIgG (Sigma, Germany)) was used as a control.
  • the cells were incubated with the incorporation of [ 3 H]-TdR (synthesized by Isotope Institute of China Institute of Atomic Energy) for 4 hours, and counted for the amount labeled by [ 3 H]-TdR using a liquid scintillation counter LKB1219 (LKB Corporation, Sweden). As shown in FIG. 12 , the result indicated that compared with the control group, the proliferation of tumor cells was inhibited significantly in the antibody BCMab1-treated group.
  • BCMab1 could significantly inhibit the growth of bladder tumors, colony formation and migration.
  • a xenograft tumor model was established by injecting T24 cells (1 ⁇ 10 7 , suspended in PBS) into the backs of BALB/c nude mice.
  • T24 cells (1 ⁇ 10 7 , suspended in PBS)
  • BCMab1 or control mIgG antibody (10 mg/kg) was injected intraperitoneally, 3 times/week.
  • tumors were reduced significantly in the BCMab1-treated group ( FIG. 12 , a).
  • the inhibition ratio reached 77% by the end of the experiment (Day 40).
  • GFP-T24 GFP fluorescence labeled T24 cells
  • IVIS in vivo imaging system
  • BCMab1 or mIgG 1 mg/kg was injected into bladders ( FIG. 12 , b).
  • administration of BCMab1 in bladder effectively inhibited growth of bladder cancer and extended lift time of T24 tumor-bearing mice (p ⁇ 0.0001) ( FIG.
  • Immunotoxin BCMab1-Ra was generated through the conjugation between BCMab1 and ricin A chain. More specifically, ricin A chain and BCMab1 were connected via a heterogeneous difunctional crosslinking agent SPDP (N-succinimidyl 3-(2-pyridyldithiol)propionate) into the immunotoxin BCMab1-Ra. The detailed procedure was shown as follows.
  • BCMab1-Ra was a very effective biological agent capable of killing bladder cancer cell lines (EJ and T24). BCMab1-Ra could inhibit the proliferation and growth of human bladder cancer cell lines in vitro (data not shown). Interestingly, direct infusion of BCMab1-Ra into bladders through urethral injection could inhibit tumor growth in mouse orthotopic transplantation tumor 1 to model which had the same survival rate as normal mice ( FIG. 12 , b-d). No toxicity caused by BCMab1-Ra was observed. As a result, BCMab1-Ra can be used as a potential toxic medicament for the treatment of bladder cancer.
  • MVEGF microvessel density
  • cytokines such as VEGF and IL-6
  • FIG. 12 , f the number of MVD was reduced significantly
  • FIG. 12 , g, h the number of MVD was reduced significantly
  • FIG. 12 , g, h the number of MVD was reduced significantly
  • FIG. 12 , g, h the number of MVD was reduced significantly
  • FIG. 12 , g, h generation of VEGF and IL-6 was inhibited in tumors.
  • BCMab1-Ra treatment group same results were shown in the BCMab1-Ra treatment group as well (data not shown). It was suggested that BCMab1 could also suppress angiogenesis while inhibiting proliferation of tumors.
  • BCMab1 could cause stasis of tumor cell cycles by preventing focal adhesin kinase (FAK) and c-Jun from phosphorylation ( FIG. 16 , a, b).
  • FAK focal adhesin kinase
  • c-Jun from phosphorylation
  • FIG. 16 , a, b The glycosylated ⁇ 3 ⁇ 1 on T24 cells enhanced FAK-dependent signalings.
  • Knockdown of integrin ⁇ 3 or GALNT1 lowered the phosphorylation levels of FAK and c-Jun significantly.
  • BCMab1 still could inhibit growth of cancer ( FIG. 16 , d). All these results suggested that BCMab1 could inhibit tumor growth and improve the killing function of NK cells.
  • T24 cells were starved in serum-free RPMI-1640 for 24 hours, and then cultured in RPMI-1640 containing 10% FBS and 1 ⁇ g/ml BCMab1 or mIgG control. After 48 hours, the cells were lysed with lysis buffer. The lysate supernatant was analyzed by immunodotting with anti-human FAK, FAK, Phospho-FAK (Tyr397), c-Jun, Phospho-c-Jun (Ser63), Phospho-c-Jun (Ser73), CyclinD1 or CDK4 antibodies.
  • the antibodies used in the above experiments include: rabbit anti-human CyclinD1, rabbit anti-human CDK4, rabbit anti-human c-Jun, Phospho-c-Jun (Ser63), Phospho-c-Jun (Ser73), rabbit anti-human FAK, Phospho-FAK (Tyr397) (CST, US), and homotypical control mIgG (Sigma, Germany).
  • Mouse monoclonal antibody BCMab1 was isolated from ascites using Protein A Sepharose.
  • the biotinylatd HRP and FITC labeled secondary antibodies were purchased from Pierce, US and Sigma, Germany, respectively.
  • Integrins impacted the adhesion-related effects in tumor cells, including proliferation, survival, migration, invasion and passage of FAK signal pathway. Phosphorylation of FAK was deemed as one of the major steps realizing further phosphorylation of Src and Fyn and triggering of signal pathway. Glycosylation of integrin ⁇ 3 ⁇ 1 was clearly different in tumor cell lines and in the control cell line. The connection between cells and extracellular matrix protein was modulated by glycosylation. We found that knockdown of integrin ⁇ 3 or GALNT1 could inhibit proliferation of cancer and growth of tumors in vivo and in vitro. Suppression of integrin ⁇ 3 ⁇ 1 signaling by BCMab1 also could inhibit proliferation of cancer and growth of tumors in vivo and in vitro.
  • BCMab1 treatment downregulated phosphorylation of FAK and c-Jun, reduced expression level of cyclin D1 and CDK4, and interfered with cell proliferation and cell cycles. Therefore, aberrantly glycosylated integrin ⁇ 3 ⁇ 1 might result in malignant transformation of cells.
  • BCMab1 could suppress tumor growth by inhibiting angiogenesis in the microenvironment of bladder cancer tumor.
  • BCMab1 downregulated the generation of VEGF and IL-6 in tumors, which suggested that targeting of tumor microenvironment required further investigation.
  • Integrin ⁇ 3 ⁇ 1 modified by abnormal glycosylation is intimately correlated with clinical pathology of bladder cancer tissues Expression of aberrantly glycosylated Integrin ⁇ 3 ⁇ 1 Total Patients High Low P Age Mean 62.3 65.7 58.9 0.352 Sex Male 48 25 23 0.732 Female 21 10 11 Tumor stage Ta, Tis, T1 26 5 21 ⁇ 0.001 T2 15 9 6 T3 20 13 7 T4 8 5 3 Grade G1 or 2 22 5 17 ⁇ 0.001 G3 47 32 15 Configuration Papillary 17 6 11 0.108 Nonpapillary 52 30 22 Number of tumors Solitary 38 23 15 0.071 Multiple 31 12 19 Lymphatic invasion Negative 18 2 16 ⁇ 0.005 Positive 39 21 18 Unknown 12 7 5
  • Follow-up (month) Mean 72.6 70.3 74.9 0.478
  • Bladder cancer is a common disease with an incidence increasing year after year. More and more evidence demonstrated that molecular signal pathway affected cell stability and thus caused bladder cancer.
  • Cell growth mainly involves five aspects, including cell cycle, cell death, cell growth, signal transduction, and gene regulation. Both the influence of external carcinogenic signals or variation of genetic factors may result in signal disorder. Maintenance and progress of tumors depend on two external factors, i.e. the correlations between interstitial substance and knizocyte, and between angiogenesis and tumor cell invasion. However, the molecular mechanism for malignant transformation of bladder cancer was still unknown.
  • the novel monoclonal antibody BCMab1 prepared by the inventors could specifically bind to aberrantly glycosylated integrin ⁇ 3 ⁇ 1 and recognize aberrantly glycosylated ⁇ 3 subunit.
  • High expression of glycosyltransferase GALNT1 may result in aberrant glycosylation of integrin ⁇ 3.
  • Knockdown of integrin ⁇ 3 or GALNT1 could inhibit proliferation of cancers and growth of tumors in vivo and in vitro.
  • block of integrin ⁇ 3 signal by BCMab1 could also inhibit proliferation of cancers and growth of tumors.
  • aberrant glycosylation level of integrin ⁇ 3 ⁇ 1 was correlated with clinical staging and prognosis of bladder cancer.
  • a hypersensitized competitive ELISA method is developed in the invention to detect bladder tumors in human urine.
  • the major process includes steps of using antigen AG- ⁇ 3 ⁇ 1 as an immobile phase and standard sample; using antibody BCMab1 as a detection antibody in combination with the secondary antibody (horseradish peroxidase (HRP) labeled goat-anti-mouse IgG); and detecting bladder cancer antigen AG- ⁇ 3 ⁇ 1 in urine of normal human and bladder cancer patients.
  • HRP horseradish peroxidase
  • the immunodiagnosis reagent for bladder cancer according to the invention had, compared with prior art, the following positive effects: (1) it directly detects urine liquid without injury, so as to avoid inconvenience and distress caused to patients by cystoscopy; (2) it has a high sensitivity and specificity that are much higher than those of the detection of current bladder tumor marker (such as nuclear matrix protein, cytokeratin, hyaluronic acid and the like) and pathological examination of urine exfoliative cells; (3) it has a high specificity and sensitivity and utilizes competitive ELISA to detect bladder cancer antigen in cells, so as to avoid the defects of higher factitiousness and lower sensitivity which cannot be overcome by cell pathology itself; and (4) it is convenient and fast, thereby suitable for screening early-stage bladder cancer patients and detecting relapse of tumors.
  • current bladder tumor marker such as nuclear matrix protein, cytokeratin, hyaluronic acid and the like
  • Clinical sensitivity can be used to evaluate the capability of a certain assay to detect sick subjects, and sensitivity is the proportion of true positives correctly identified in the actual sick subjects.
  • Clinical specificity can be used to evaluate the capability of a certain assay to identify healthy subjects, and specificity is the proportion of true negatives correctly identified in the actual healthy subjects.
  • T24 cells endogenously expressing GAL3ST2 protein, were taken and cultured as above.
  • the cells were used as cell models to screen for the medicaments to treat bladder cancer in mammals.
  • Test group the above cell culture was treated with a candidate
  • Control group the above cell culture was not treated with the candidate.
  • the cells were detected for GAL3ST2 expression, enzyme activities, existing amount or secretion status by 1 to conventional methods.
  • GAL3ST2 protein expression, enzyme activities, existing amount or secretion was significantly reduced by 30% or more in the test group, which indicated that such a candidate is a potential substance to treat bladder cancer.
  • RNAi sequence of GAL3ST2 was tested as a candidate. The result showed that GAL3ST2 protein expression was inhibited. Thus the RNAi sequence is a potential substance to treat bladder cancer.
  • T24 cells endogenously expressing N-acetylgalactosaminyltransferase 1 protein, were taken and cultured as above. The cells were used as cell models to screen for the medicaments to treat bladder cancer in mammals.
  • Test group the above cell culture was treated with a candidate
  • Control group the above cell culture was not treated with the candidate.
  • the cells were detected for N-acetylgalactosaminyltransferase 1 expression, enzyme activities, existing amount or secretion status by conventional methods.
  • N-acetylgalactosaminyltransferase 1 protein expression, enzyme activities, existing amount or secretion was significantly reduced by 30% or more in the test group, which indicated that such a candidate is a potential substance to treat bladder cancer.
  • RNAi sequence of N-acetylgalactosaminyltransferase 1 was tested as a candidate. The result showed that N-acetylgalactosaminyltransferase 1 protein expression was inhibited.
  • the RNAi sequence is a potential substance to treat bladder cancer.

Abstract

Provided in the present invention are an aberrantly glycosylated integrin, AG-α3β1, and use thereof as a bladder cancer marker. Also provided in the present invention are a hybridoma cell generating an anti-AG-α3β1 monoclonal antibody, a monoclonal antibody BCMab1 secreted by the same, and use of BCMab1 in the preparation of a medicament for the treatment of bladder cancer. Also provided in the present invention is use of inhibitors of GAL3ST2 and N-acetylgalactosaminyltransferase 1 in the preparation of a medicament for the treatment of bladder cancer.

Description

    FIELD OF THE ART
  • The present invention belongs to a field of tumor immunology. Particularly, the present invention relates to a novel antigen AG-α3β1 and the preparation thereof (AG: Aberrant Glycosylation), and an anti-AG-α3β1 monoclonal antibody BCMab1. It is demonstrated at cytology and histology levels that AG-α3β1 is expressed on the membranes of human bladder tumor cells only, the antibody BCMab1 not only specifically recognizes AG-α3β1, but also effectively inhibits the proliferation of human bladder tumor cells at cellular level in vitro and in an animal model in vivo. The present invention also relates to a process for detecting bladder cancer in human using a competitive ELISA.
  • In such a detecting process, the antigen in an immobile phase is human bladder cancer tumor marker AG-α3β1 according to the present invention, and the detection antibody is the antibody BCMab1 directed against human bladder cancer integrin α3β1.
  • BACKGROUND OF THE INVENTION
  • Bladder cancer is a tumor most commonly found in urinary systems. Although numerous steps were adopted for the treatment, 40-70% of the patients relapsed once or more than once and 10-15% of the patients developed more severe tumors or involved metastasis. Moreover, all these treating processes have apparent toxic and side-effects.
  • One important clinical task is to prevent relapse of bladder cancer after excision of tumors. Currently, the medicaments used for preventing relapse by bladder perfusion are mainly divided into the following several classes: (1) anti-tumor chemotherapeutic agents, such as Mitomycin C, etc.; (2) immunopotentiators, such as Bacillus Calmette-Guerin Vaccine (BCG), etc.; and (3) cytokines, such as interferon, etc. Although the above classes of medicaments, used either alone or in combination, achieved some effects in the reduction of the relapse rate after the operation of bladder cancer, the overall therapeutical effect is not desirable due to the presence of many issues such as low specificity, multiple drug resistance (MDR) of tumors, and so on. Moreover, because of their low molecular weights, the above classes of medicaments not only can act on the entire bladder and urethra unspecifically, but also can be absorbed by the mucous membranes of bladder and urethra, which easily results in topical or systematic toxic and side-effects. For instance, in the case of Mitomycin C and Bacillus Calmette-Guerin Vaccine which have more positive therapeutical effects, 90% of the patients who had received Bacillus Calmette-Guerin Vaccine were suffered from BCG cystitis and other side effects including hematuria, tetter, fever, arthritis, urethral stricture and the like, or infrequently, even some severe conditions, such as hepatitis, pneumonia, life-threatened septicemia and so on. Mitomycin C has fewer side effects, however still 5-25% of the patients who had received Mitomycin C were suffered from chemical cystitis and anaphylaxis, and other side effects included urethral stricture, marrow depression, calcification of bladder wall, and so on.
  • Integrin is a group of divalent cation-dependent cell surface receptors with a major function of mediating cell-matrix, cell-cell adhesion by binding to corresponding ligands, so as to further affect cell shapes, gene expression and regulation, cell proliferation and differentiation, apoptosis, and migration, infiltration and metastasis of tumor cells, etc. Integrin is composed of non-covalent subunits α and β. Various combinations result in different ligand-binding abilities. Integrin subunit α3 (CD49c) can be combined with subunit β1 (CD29) to form integrin α3β1. The ligands for integrin α3β1 are laminin (LN) and Collagen IV. In epithelial tissues, integrin α3β1 is mainly involved in the conglutination between cells and basilar membranes of epithelial cells and can adjust signal transduction, thereby impacting the biological features of cells. Integrin α3β1 is modified aberrantly on the surface of tumor cells, which is considered to cause a change in malignancy.
  • Anti-tumor targeted drugs are biological or chemical molecules (e.g. monoclonal antibodies) with an ability to recognize and kill tumor cells specifically. Bladder cancer is a tumor inside the body cavity of human, i.e. a “somatic test tube” of human body. Targeted drugs have high specificities with and strong lethal effects on target cells in vitro. Therefore, in terms of the treatment for bladder cancer, it is very important to find a novel targeted drug directed against bladder cancer in human.
  • In screening for bladder tumors, early detection and accurate prognosis of bladder cancer appear to be extremely critical for clinical treatment. For the past several years, the molecular markers, such as nuclear matrix protein, bladder tumor antigen and satellite instability etc. newly involved in clinic were also limited by their low sensitivities and specificities. Urine cytological analysis and cystoscopy are still the golden criteria for the diagnosis and monitoring of bladder cancer. Therefore, it is extremely necessary to find an effective and convenient process for diagnosing bladder cancer.
  • DISCLOSURE OF THE INVENTION
  • The inventors utilized human bladder cancer cell line T24 to immunize mice, so as to achieve hybridoma cells. ELISA was conducted to screen for antibody BCMab1 which can bind to T24 cells with a high specificity. It was demonstrated by immunohistochemistry and immunofluorescence that antibody BCMab1 showed a strong positive reactivity with bladder cancer cell line T24 and bladder cancer tissue of human, but no cross reactivity with normal bladder tissue or other non-bladder cancer cells.
  • In the present invention, the antigen recognized by antibody BCMab1 was captured by antibody BCMab1 using an immunoaffinity chromatography, and then identified as integrin α3β1 modified by aberrant glycosylation using a mass spectroscopy and carbohydrate chips. Such an antigen is designated as AG-α3β1, having an epitope in a carbohydrate structure shown as follow: [3OSO3]Galβ1-4(Fucα1-3)[6OSO3]GlcNAc. It has been demonstrated by the antibody BCMab1 that AG-α3β1 is only expressed on bladder tumor cells of human, and correlated positively with tumor staging and pathological grading of bladder cancer. AG-α3β1 can be used as a novel marker for bladder cancer tumors of human.
  • In the invention, T24 cells cultured in vitro and a nude mouse transplantation tumor model are treated with antibody BCMab1. It is found that antibody BCMab1 can effectively inhibit the proliferation of bladder cancer cell line T24. Antibody BCMab1 thus can be used as the targeted drug directed against bladder cancer of human.
  • In the invention, a diagnostic reagent is made from antibody BCMab1 and antigen AG-α3β1. Particularly, human bladder cancer tumor marker AG-α3β1, as an immobile phase, competes with a sample to be tested for antibody AbBC1, so as to detect bladder tumor cells in human urine.
  • It is also found in the invention that glycosyltransferase GALNT1 is highly expressed in bladder cancer and GALNT1-knockdown cells can down-regulate glycosylation level of integrin α3β1. Aberrant glycosylation of integrin α3 facilitates malignant transformation of bladder cancer. BCMab1 and immunotoxin BCMab1-Ra can significantly inhibit growth of the bladder cancer transplanted subcutaneously and orthotopically. BCMab1 treatment can recruit and activate natural killer (NK) cells surrounding bladder cancer. Both aberrant glycosylation of integrin α3β1 and severity of bladder cancer patients are associated with prognosis. As a result, aberrant glycosylation of integrin α3β1 can be considered as a new target for the diagnosis, prognosis and immunotherapy of bladder cancer.
  • Some creative ideas are brought up in the invention, including (1) preparing a mouse monoclonal antibody BCMab1 directed against human bladder cancer, which not only specifically binds to bladder cancer tissues of human, but also effectively inhibits proliferation of bladder cancer cell line T24 in vivo or in vitro; (2) revealing that the antigenic epitope AG-α3β1 recognized by such an antibody is a novel marker for human bladder cancer tumors, and demonstrating that such an epitope is only expressed on bladder tumor cells of human and correlated positively with tumor staging and pathological grading of bladder cancer; and (3) developing a highly sensitive competitive ELISA method for detecting human bladder cancer.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The invention provides an aberrantly glycosylated integrin AG-α3β1, as well as a hybridoma cell generating a monoclonal antibody directed against such a tumor marker and the monoclonal antibody BCMab1 secreted by the same. The antigenic epitope recognized by the antibody BCMab1 is [3OSO3]Galβ1-4(Fucα1-3)[6OSO3]GlcNAc. Such a monoclonal antibody shows a strong positive reactivity with human bladder cancer cell line T24 and human bladder cancer tissues, but no cross reactivity with normal bladder tissue or other non-bladder cancer cells of human. In addition, the monoclonal antibody has a function of inhibiting proliferation of bladder cancer cell line T24 in cell cultures in vitro and in animal tumor models. The invention also provides an in vitro diagnostic kit comprising the monoclonal antibody BCMab1, and a process for detecting the content of the tumor marker in urine exfoliative cells using the monoclonal antibody BCMab1.
  • More specifically, one object of the invention is to provide an aberrantly glycosylated integrin AG-α3β1 characterized in that the integrin α3β1 has a carbohydrate structure: [3OSO3]Galβ1-4(Fucα1-3)[6OSO3]GlcNAc as an antigenic epitope. The carbohydrate structure [3OSO3]Galβ1-4(Fucα1-3)[6OSO3]GlcNAc is represented by a structural formula shown as follow:
  • Figure US20130164216A1-20130627-C00001
  • In a preferred embodiment of the invention, the integrin α3β1 has an α3 subunit having an amino acid sequence shown as SEQ ID No: 1 and a β1 subunit having an amino acid sequence shown as SEQ ID No: 2. Preferably, the aberrant glycosylation is located on the amino acid T (threonine) at position 740 of the α3 subunit.
  • Another object of the invention is to provide a binding molecule directed against the AG-α3β1 according to the invention. Such a binding molecule can specifically recognize or bind to the carbohydrate structure [3OSO3]Galβ1-4(Fucα1-3)[6OSO3]GlcNAc as the antigenic epitope. Preferably, the binding molecule is a polyclonal antibody or monoclonal antibody. More preferably, the binding molecule is a monoclonal antibody.
  • In a preferred embodiment, the binding molecule is an anti-AG-α3β1 monoclonal antibody BCMab1, and the monoclonal antibody is secreted by the hybridoma cell line deposited as CGMCC No. 3845.
  • Another object of the invention is to provide a conjugate, wherein the binding molecule of the invention is conjugated with a substance selected from the group consisting of a biological marker, an antitumor drug, a toxin and a radioactive agent. Preferably, the conjugate is an immunotoxin BCMab1-Ra prepared from BCMab1 and ricin A chain connected by a heterogeneous difunctional crosslinker such as SPDP (N-succinimidyl 3-(2-pyridyldithio) propionate), MBS (m-Maleimidobenzoyl-N-hydroxysuccinimide ester), or 2-IT (2-Iminothiolane), etc. The connection method between an antibody and ricin A chain is well known and can be found in the following references for details: Shen, B. Connection between monoclonal antibody and toxin, Monoclonal Antibody Communication 1989; 2: 51; Cumber A J, et al. Preparation of antibodytoxin conjugates, Methods Enzymol 1985; 112:207; and Cros o, et al. Biochemical aspect of immunotoxin preparation, J. Immunol. Meth. 1985; 81:283.
  • Another object of the invention is to provide a pharmaceutical composition which comprises an effective dose of the binding molecule according to claims 3 or 4 or the conjugate according to claim 5, and a pharmaceutically acceptable carrier, diluents or excipient.
  • A further object of the invention is to provide a kit for detecting bladder cancer, which comprises the binding molecule of the invention, the conjugate of the invention, or AG-α3β1; and optionally another reagent for detecting bladder cancer. In a preferred embodiment, the detection is carried out by an enzyme linked immunosorbent assay. Preferably, the enzyme linked immunosorbent assay is a competitive enzyme linked immunosorbent assay, wherein AG-α3β1 of the invention, as an immobile phase, competes with a sample to be tested for the antibody of the invention.
  • In a preferred embodiment, the sample to be tested in said kit is urine or bladder tissue.
  • Another object of the invention is to provide a hybridoma cell line secreting the antibody BCMab1 directed against integrin α3β1 of human bladder cancer, which is deposited as CGMCC No. 3845.
  • A further object of the invention is to provide use of the pharmaceutical composition of the invention in preparation of the medicament for the treatment of bladder cancer. In a preferred embodiment, the medicament is used in a combination therapy with other drugs for the treatment of bladder cancer. Said other drugs for the treatment of bladder cancer are preferably selected from the group consisting of antitumor chemotherapeutic agent, immunoreinforcing agent, and cytokines; and more preferably a group consisting of thiotepa (TSPAP), Mitomycin C (MMC), Bacillus Calmette-Guerin Vaccine (BCG), cisplatin (DDP), adriamycin (ADM) and interferon. Further more preferably, said medicament and said other drugs for the treatment of bladder cancer are used simultaneously, separately or sequentially for the treatment of human or animal bodies. Still preferably, said medicament and said other drugs for the treatment of bladder cancer, formulated in a same or different preparations, are co-administered to the subjects in need thereof through a same or different routes.
  • Another object of the invention is to provide use of the AG-α3β1 or antibody of the invention, optionally combined with other indicators for the diagnosis or prognosis of bladder cancer, in the diagnosis and/or prognosis of bladder cancer.
  • Another object of the invention is to provide use of the AG-α3β1 of the invention for preparing and screening antibodies, wherein the AG-α3β1 is used as an immunogen to immunize animals, so as to obtain monoclonal or polyclonal antibodies, or the AG-α3β1 is used as an antigen to screen and prepare antibodies by a phage, yeast or ribosome display technique. Another object of the invention is to provide use of an RNAi sequence directed against GAL3ST2 in preparation of the medicament for the treatment of bladder cancer. Preferably, the nucleotide sequence of the RNAi is shown as SEQ ID No: 3.
  • Another object of the invention is to provide use of an inhibitor of N-acetylgalactosaminyltransferase 1 in preparation of the medicament for the treatment of bladder cancer. Preferably, the inhibitor includes the nucleotide sequence of the RNAi as shown by SEQ ID No: 4, uridine 5′-phosphoric (1-hexadecanesulfonic) anhydride, and Ammonium (3-Acetamido-2,6-anhydro-3-deoxy-D-glycero-L-gluco-heptitol-1-yl) (Uridine-5′-yl)Diphosphate.
  • Another object of the invention is to provide a process of screening for a potential substance to treat bladder cancer, which comprises the steps of:
      • (1) treating a system expressing N-acetylgalactosaminyltransferase 1 protein or GAL3 ST2 protein with a candidate; and
      • (2) detecting the expression or activity of the N-acetylgalactosaminyltransferase 1 protein or GAL3ST2 protein in the system;
        wherein, if the candidate can reduce the expression or activity of the N-acetylgalactosaminyltransferase 1 protein or GAL3ST2 protein, then the candidate is indicated to be a potential substance to treat bladder cancer.
  • Yet another object of the invention is to provide use of N-acetylgalactosaminyltransferase 1 in the diagnosis and/or prognosis of bladder cancer.
  • DESCRIPTION OF THE FIGURES
  • FIG. 1. BCMab1 specifically recognized bladder cancer cell line and bladder cancer tissue. (a, b) It was shown by immunofluorescence and immunohistochemisty that BCMab1 stain was positive for bladder cancer cell line T24 cells and negative for normal bladder cancer cell line HCV29. (c) BCMab1 stain was positive for bladder cancer tissues and negative for normal bladder mucosal tissues (IHC);
  • FIG. 2. Periodide-Oxidation Experiment (Experimental principle of the Periodic acid-Oxidation experiment: a hydroxyl group of the carbohydrate in glycoprotein is oxidized by a periodic acid in an acidic condition without any alteration of the polypeptide chain structure). In FIG. 2, bladder cancer cells T24 were treated with iodic acid to destroy the carbohydrate antigen structure, so that antibody BCMab1 cannot recognize and no positive reaction with a brown color was present. Anti-MUC1 antibody (commercially purchased from Novocastra Laboratories Ltd, UK) is an antibody known to bind to carbohydrate structures only, which was used as a control herein. Anti-CK20 antibody (Novocastra Laboratories Ltd, UK) is an antibody known to bind to proteinic polypeptides only, which was also used as a control herein;
  • FIG. 3. Periodide-Oxidation Experiment (Dot blot hybridization experiment, with a result similar with FIG. 2). After lysis, bladder cancer cells T24 were spotted on a film and treated with iodic acid, so that the carbohydrate antigen structure was destroyed, carbohydrate antigen structure was not able to be detected by the antibody BCMab1 and no positive reaction was present;
  • FIG. 4. Results of the carbohydrate chip assay;
  • FIG. 5. Knockdown of integrin α3 or integrin β1 in T24 cells (Vector is a vector control, and β-actin is an internal standard for amplification);
  • FIG. 6. Integrin α3-knockdown T24 cells were barely stained by BCMab1, whereas integrin β1-knockdown T24 cells were stained (shIntegrin α3 and shIntegrin β1 represent the cells in which the α3 and β1 subunits are knocked down by RNAi, respectively);
  • FIG. 7. Impact of GAL3ST2 on aberrant glycosylation of integrin α3β1 in bladder cancer tissues. (a) Puro-screening for GAL3ST1-4-silenced T24 cells and RT-PCR detection for the expression level. (b) BCMab1 barely stained GAL3ST2-knockdown cells. (c) Compared with the control cells transfected with an empty vector, knockdown of GAL3ST2 could reduce the proliferation of T24 cells significantly. (d) Knockdown of GAL3ST2 could reduce the colony forming ability of T24 cells significantly. (e) Knockdown of GAL3ST2 could reduce the adhesion of T24 cells significantly. (f, g) Knockdown of GAL3ST2 could reduce cell migration significantly;
  • FIG. 8. Quantitative RT-PCR detection for GALNT1 silence effect in EJ cells;
  • FIG. 9. GALNT1 silence almost blocked BCMab1 staining. T24 cells and an inhibitor for GALNT1, i.e. BG were co-incubated for 7 days, and then stained with BCMab1;
  • FIG. 10. Knockdown of integrin α3 or GALNT1 inhibited cancer cell proliferation and tumor growth. (a) Silence of integrin α3 or GALNT1 inhibited proliferation of T24 cells. Integrin α3- or GALNT1-knockdown T24 cells or BCMab1 (10 μg/ml)-treated T24 cells, after cultured for 24-72 hours, were detected for cell proliferation by 3H-TdR incorporation assay. (b) Integrin α3- or GALNT1-silenced cell lines could not grow in a soft agar. After a treatment with BCMab1 (10 μg/ml), the growth of T24 cells was also inhibited in a soft agar. (c) Silence of integrin α3 or GALNT1 inhibited growth of bladder cancer. T24 cells transfected with integrin α3 or GALNT1 were injected subcutaneously into back of a BALB/c mouse. The tumor sizes were measured periodically, and averaged (the left panel). The right panel shows the tumor tissues;
  • FIG. 11. Knockdown of integrin α3 inhibited the adhesion between tumor and extracellular matrix (ECM) and tumor migration, as well as facilitated NK cell-mediated cell killing. (a, b) Compared with an empty vector, knockdown of integrin α3 weakened the adhesion between tumors and ECM (a) and tumor migration (b) (p<0.01); whereas knockdown of GALNT1 failed to show an evident result. BCMab1 (10 μg/ml) could inhibit cell junction and tumor migration significantly (mIgG as negative control). (c) It was confirmed in a scratch test that knockdown of integrin α3 could inhibit migration of tumor cells (p<0.01). (d) Silence of integrin α3 or GALNT1 could improve the cell killing caused by NK cells. 51Cr-labeled integrin α3 or GALNT1-silenced EJ cells and IL-2-activated NK cells (left) or NK92 cells (right) were incubated together at 37° C. for 4 hours in various effect-target ratios (E/T). (e) Integrin α3 or GALNT1-knockdown cells were inoculated into the mice from which NK cells had been removed, and the growth of tumor cells was accelerated. NK cells were removed using anti-asialo GM (GM) antibody, and then the integrin α3 or GALNT1-knockdown EJ cells were inoculated. Tumor volumes were measured periodically, and averaged (the left panel). The tumor status on Day 35 is shown in the right panel. (f) Removal of NK cells impacted tumors. After a TUNEL staining (left), the average numbers of the positively stained cells were calculated (the right panel). (g, h) Tumors inoculated with integrin α3-/GALNT1-knockdown EJ cells or BCMab1-treated EJ cells, compared with tumors inoculated with EJ cells transfected with an empty vector or mIgG-treated EJ cells, could significantly facilitate the secretion of IFNγ and perforin by NK cells. IFNγ was detected by ELISA and perforin was detected by FACS. (i) Tumors inoculated with integrin α3-/GALNT1-knockdown EJ cells or BCMab1-treated EJ cells, compared with tumors inoculated with EJ cells transfected with an empty vector or mIgG-treated EJ cells, could facilitate the recruitment of NK cells inside tumors;
  • FIG. 12. Antibody BCMab1 inhibited the proliferation of tumor cells;
  • FIG. 13. Amino acid sequence of integrin α3 subunit;
  • FIG. 14. Amino acid sequence of integrin β1 subunit;
  • FIG. 15. BCMab1 and BCMab1-Ra inhibited growth of the transplanted tumors of a mouse-borne tumor model. (a) In a xenografted mouse-borne tumor model, BCMab1 and BCMab1-Ra inhibited cancer growth. T24 cells were injected into nude mice subcutaneously. When the tumor diameter reached 3-5 mm, the mice were grouped (10 mice/group), and injected with BCMab1 or BCMab1-Ra (10 mg/kg) intraperitoneally, 3 times/week, for 5 weeks. The tumor statue on Day 35 is shown in the lower panel. (b) The tumor-suppression effect of BCMab1 and BCMab1-Ra on orthotopically transplanted bladder cancer. After transplanted, GFP-labeled T24 cells were imaged by IVIS on Days 5, 10, 15, 20 and 25. Chromatic aberration showed photon amounts of the mice. Groups of mice (24 mice/group) were administered by infusing bladder, and then given with BCMab1, BCMab1-Ra and mIgG (1 mg/kg each) respectively on the next day. (c) The growth curve of the orthotopic tumor after IVIS treatment. (d) Comparison of the growth curves for BCMab1, BCMab1-Ra and mIgG treated groups. (e) It was shown by immunohistochemisty and HE staining that injection of BCMab1 and BCMab1-Ra in bladder successfully inhibited tumor growth. The tumor was taken to a histological analysis on Day 25 during the treatment. No tumor cell was detected in BCMab1-Ra-treated mice. (f) BCMab1 downregulated the microvessel density (MVD) in tumors. BCMab1 and mIgG-treated mice were subjected to an immunohistochemistry assay using anti-CD31 antibody on Days 10, 20 and 30, so as to calculate MVD (measure microvessel density) (the right panel). (g, h) ELISA results showed that levels of VEGF and IL-6 in tumors were downregulated significantly in the BCMab1-treated mice;
  • FIG. 16. BCMab1 resulted in cell cycle arrest and inhibited tumor growth. (a) BCMab1 resulted in cell cycle arrest. BCMab1 (10 μg/ml)-treated T24 cells were analyzed by a PI (Propidium Iodide)-staining flow cytometer. mIgG was used as a negative control. (b) Antibody BCMab1 inhibited phosphorylation of FAK and c-Jun, and downregulated expression of cyclin D1 and CDK4. The lysate was immunoblotted by anit-phospho-FAK (Tyr397) and anti-phospho-c-Jun (Ser63 and Ser73) as well as the total FAK and c-Jun. At the same time, the expression of cyclin D1 and CDK4 was detected. β-actin was used a control. (c) Aberrant glycosylation of integrin α3β1 enhanced the activation of FAK signal pathway. T24 cells and integrin α3- or GALNT1-silenced T24 cells were treated with Matrigel in a serum-free condition for 30 min. HCV29 cells were used as a normal control. (d) In NK cell-absent mice, BCMab1 still could inhibit tumor growth. After removal of NK cells using anti-asialo GM1 (GM1) antibody, the mice were administered with BCMab1, or inoculated with Integin α3-knockdown T24 cells. On Day 35, tumor volumes were measured, and averaged (the left panel). The right panel shows the tumor tissues;
  • FIG. 17. Aberrant glycosylation of integrin α3β1 was correlated with clinical severity and prognosis. (a) Immunohistochemical results showed that aberrant glycosylation expression level of integrin α3β1 was correlated with clinical characters. The intensity of BCMab1 staining was related with pathological staging and grading. (b) According to the visits for the prognoses of bladder cancer patients in combination with immunohistochemical analysis, it was indicated that expression of integrin α3β1 was correlated with pathological characters in clinic. The expression level of integrin α3β1 was high in the bladder cancer specimen with a higher grade, deeper invasion and lymph-node involvement; whereas low in the case with a lower grade, superficial invasion and lymph-node involvement-free. The patients with high expression levels of integrin α3β1 showed poor prognoses (p=0.0036).
  • DETAILED EMBODIMENTS OF THE INVENTION
  • As used herein, the term “binding molecules” means intact immunoglobulins, including monoclonal antibodies and polyclonal antibodies, such as chimeric antibodies, humanized antibodies or human monoclonal antibodies, or antigen binding fragments which can bind to antigenic epitopes, including Fab, F(ab′), F(ab′)2, Fv, dAb, Fd, complementary determining region (CDR) fragment, single-chain antibodies (scFv), bivalent single-chain antibodies, single-chain phage antibodies, bispecific double-chain antibodies (diabodies), triabodies, tetrabodies, and proteins or polypeptides binding to antigenic epitopes, etc. The above fragments can be generated through synthesis, enzymatic or chemical lysis of intact immunoglobulins, or recombinant DNA technology-genetic engineering. All these preparation methods are well known in the art, for example, E. Harlow and D, Lane (1988) ed. Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., which is incorporated herein by reference.
  • As used herein, the term “binding molecules” include all classes and subclasses of immunoglobulins known in the art. According to the amino acid sequences of their heavy-chain constant domains, binding molecules can be divided into 5 major classes of intact antibodies: IgA, IgD, IgE, IgG and IgM, wherein some classes can be further divided into subclasses (isotypes), e.g. IgA1, IgA2, IgG1, IgG2, IgG3 and IgG4.
  • An anti-AG-α3β1 monoclonal antibody, BCMab1 is provided in the invention, and anti-AG-α3β1 polyclonal antibodies and other monoclonal antibodies can be easily generated by a person skilled in the art. The general processes for preparing a monoclonal antibody from hybridoma are well known. The cell lines that generate immortal antibodies can be prepared through cell fusion and other techniques, such as transformation of B lymphocytes by oncogenic DNA, or EB (Epstein-Barr virus) transfection.
  • Physical selection is one example of the conventional processes for obtaining all human antibodies, and includes but is not limited to screening by a phage, yeast or ribosome display technique. The detailed procedure comprises a step of selecting a ligand (antibody) binding to the target protein (antigen) accordingly. There are two conventional physical selection methods. In the first method, an antigen is fixed on a solid support and incubated with an antibody library; after recognition and binding between the antibodies and antigens, unbound antibodies are rinsed off, and the antibodies bound specifically are eluted, collected, identified and stored. In the second method, an antigen is labeled (with biotin or fluorescence, usually) and then the antibodies specifically binding to such an antigen are screened out. In phage, bacterium and yeast display techniques, a living organism is used for the amplification and display, and phenotype and genotype are combined; whereas ribosome and puromycin mRNA display techniques rely on PCR amplification, in vitro RNA translation for obtaining a ligand, and then the binding to corresponding cDNA via a noncovalent bond or a covalent bond. These methods usually require several rounds of screenings. Since the antigenic specificities are known in advance, therefore the specificities of the resultant antibodies are definite. All these methods are well known by a person skilled in the art, and described detailedly in Antibody Engineering Drugs (Zhen, Y. and Shao, R., Chemical Industry Press, published on 2002 Nov. 1).
  • The invention first reveals a relation between the increase of AG-α3β1 and development/prognosis of bladder cancer, and demonstrates that AG-α3β1 can be used for the diagnosis, therapeutic direction and prognosis. It is understood by a person skilled in the art that all kinds of methods, including but not limited to immunohistochemistry, immunoenzyme technique, enzyme linked immunosorbent assay, immunofluorescence technique, radioimmunoassay, immune-colloidal gold labeling technique and so on, can be applied to detect AG-α3β1.
  • After determination of the coating antibody and detection antibody used in the kit of the invention, all types of markers, which can be detected commonly by binding to the detection antibodies in the field, can be applied. The markers applied in the invention are not specifically limited, as long as they can bind to the detection antibodies and, after treated properly, accurately indicates whether AG-α3β1 is present in the samples to be detected and the existing amount thereof. For instance, the markers can be selected from (but not limited to): horseradish peroxidase, alkaline phosphatase, glucose oxidase, β-D-galactosidase, urease, catalase, or glucoamylase.
  • Where some of the above enzymic markers are used, some substances that bind to the corresponding enzymes are also required, so that the existing status or amounts of the markers can be reported by color development and so on. The substances include (but are not limited to): o-phenylene diamine (OPD), tetramethyl benzidine (TMB) and ABTS for horseradish peroxidase; and p-nitrophenyl phosphate (p-NPP) for alkaline phosphatase.
  • In order to eliminate false positive and false negative, a quality control (i.e. control) is intended to be set up during the detection. In one example of the invention, the quality control is prepared by diluting a standard AG-α3β1 preparation and serum obtained from healthy volunteers. In addition, if quantitative results are desired, a person skilled in the art understands that several standard AG-α3β1 preparations with known concentrations can be set up during the detection. The standard preparations can be set up by conventional methods.
  • As used herein, the “sample” means a substance extracted from human or animal bodies, and includes but is not limited to: urine, plasma, serum, bladder tissue, and other tissues or extraction liquids thereof. Preferably, the “sample” is urine. After lysis, bladder cancer cells release various types of soluble molecules, including AG-α3β1.
  • Pharmaceutical Composition and Use Thereof
  • The invention further provides a pharmaceutical composition, which comprises:
  • (a) an effective amount of the binding molecules of the invention; and a pharmaceutically acceptable carrier, diluents or excipient.
  • As used herein, the term “the composition of the invention” includes pharmaceutical compositions, as long as they comprise the binding molecules of the invention as an active component.
  • In the invention, the term “comprise” means that all types of components can be applied to the mixture or composition of the invention.
  • In the invention, a “pharmaceutically acceptable” component is a substance which is suitably used in human and/or animals without causing excessive adverse side effect (such as toxicity, irritation and allergy), i.e. a substance with a reasonable efficiency/risk ratio. As used herein, the term “an effective amount” means the amount of a therapeutical agent to treat, alleviate or prevent diseases or conditions, or the amount showing detectable therapeutical or prophylactic effect. The accurate effective amount for a particular subject depends on its somatotype and healthy condition, feature and severity of the disease, as well as the selected therapeutical agents and/or the combination thereof for administration. As a result, predetermination of the accurate effective amount is meaningless. However, in terms of some specified circumstances, the effective amount can be determined by a clinical physician through conventional experiments. For the purpose of the invention, the effective amount means administration of 0.01 μg/kg-500 μg/kg, preferably 0.05 μg/kg-200 μg/kg body weight of the active substance of the invention into individuals.
  • The pharmaceutical composition can further comprise a pharmaceutically acceptable carrier. The term “a pharmaceutically acceptable carrier” means a carrier used for administering a therapeutical agent. A pharmaceutically acceptable carrier satisfies the following criteria: it neither induces generation of the antibodies which harm the individuals who have received the composition, nor causes excessive toxicity upon administration. Such carriers are well known by a person skilled in the art. A sufficient discussion of the pharmaceutically acceptable carriers can be found in Remington's Pharmaceutical Sciences, Mack Pub. Co., N.J. 1991. Solid carriers include but are not limited to: starch, lactose, dicalcium phosphate, microcrystalline cellulose, sucrose and kaolin; and liquid carriers include but are not limited to: saline, buffer, glucose, water, glycerol, ethanol, adjuvant, polyethylene glycol, nonionic surfactant and edible oil (such as corn oil, peanut oil and sesame oil), as long as they are suitable for the properties of the active components and the desired specific administration routes. The adjuvants commonly used for preparing pharmaceutical compositions can also be included advantageously, for example, a flavoring agent, pigment, preservative and antioxidant such as vitamin E, vitamin C, BHT and BHA. Additionally, these carriers possibly further comprise auxiliaries such as a wetting agent or emulsifying agent, pH buffer substance and so on.
  • The pharmaceutical composition of the invention can be prepared into any conventional preparation forms by conventional methods.
  • The dosage forms of the pharmaceutical composition according to the invention are various, as long as they enable the active component to enter into mammalian bodies effectively. The composition can be formulated into any forms suitable for the desired administration routes. For instance, the pharmaceutical composition can be made into tablets, pills, powder, lozenges, pouches, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosol (as a solid or in a liquid matrix), ointment, soft and hard gel capsules, suppositories, aseptic injections, aseptic packagings of powder, and so on.
  • The binding molecules or their pharmaceutically acceptable derivatives, modifications, and combinations thereof can be administered through bladder infusion, intravenous, intramuscular or subcutaneous injection, and an oral route, etc., preferably through bladder infusion. The composition of the invention and the pharmaceutical composition thereof can also be stored in a disinfector suitable for injection or dripping.
  • The composition of the invention, once prepared, can be given to a subject directly. The subjects to be prevented or treated can be animals; particularly human. The therapeutical dose regimen can be a single-dose regimen or a multidose regimen, which can be adjusted for providing an optimal therapeutical response. For example, due to the stringent requirements of the therapeutical status, several separated doses can be given every day, or the dose can be reduced proportionally.
  • The composition or medicament of the invention can further comprise other active components, e.g. other drugs for the treatment of bladder cancer used in a combination therapy. Said other drugs for the treatment of bladder cancer are selected from preferably the group consisting of antitumor chemotherapeutic agent, immunoreinforcing agent, and cytokines; and more preferably a group consisting of thiotepa (TSPAP), Mitomycin C (MMC), Bacillus Calmette-Guerin Vaccine (BCG), cisplatin (DDP), adriamycin (ADM) and interferon. Where two or more drugs mentioned above are administered in a combination, normally an effect that excels those caused by administering the two drugs separately is achieved. Preferably, the drugs or other preparations administered in a combination do not interfere with the therapeutical activity of the binding molecules according to the invention.
  • It was found in the invention that knockout of N-acetylgalactosaminyltransferase 1 (GalNAc-T1) expression could inhibit the proliferation of bladder cancer cells and the growth of tumors. Thus a person skilled in the art understands that all inhibitors for GALNT1 can be applied to prepare the drugs for the treatment of bladder cancer. The inhibitors include but are not limited to the nucleotide sequence (SEQ ID No: 4) of RNAi mentioned in Examples, an inhibitor for GALNT1, i.e. ammonium (3-Acetamido-2,6-anhydro-3-deoxy-D-glycero-L-gluco-heptitol-1-yl) (Uridine-5′-yl)Diphosphate disclosed by Thiem J. (see the following references: J Org. Chem. 2000 Jan. 14; 65(1):24-9. Synthesis of novel donor mimetics of UDP-Gal, UDP-GlcNAc, and UDP-GalNAc as potential transferase inhibitors.), uridine 5′-phosphoric (1-hexadecanesulfonic) anhydride disclosed by Elbein A D (see the following reference: Biochem Biophys Res Commun. 1991 Mar. 15; 175(2):668-72. Synthesis of a new inhibitor of the UDP-GalNAc: polypeptide galactosaminyl transferase.), a inhibitor for GalNAc-T1 disclosed in WO 2010/118347 A2, and the like.
  • It was also found in the invention that GALNT1 mRNA expression level was higher in bladder cancer tumor tissues than in normal tissues. Therefore, GALNT1 mRNA can be used as indicator for diagnosis and/or prognosis of bladder cancer. Conventional methods for detecting GALNT1 mRNA are known by a person skilled in the art. Specifically, the tissue to be detected and the normal control tissue are obtained, and detected for their GALNT1 mRNA contents; if the GALNT1 mRNA level is higher in the tissue to be detected than in the normal control tissue by 2 folds or more, then the tissue to be detected is indicated to be a bladder cancer tissue.
  • It was further found in the invention that knockout of GAL3ST2 (galactose: 3-O-sulfonyltransferase 2) expression inhibited the proliferation of bladder cancer cells and the growth of tumors. Thus a person skilled in the art understands that all inhibitors for GAL3ST2 can be applied to prepare the drugs for the treatment of bladder cancer. The inhibitors include but are not limited to the nucleotide sequence (SEQ ID No: 3) of RNAi mentioned in Examples and an RNAi inhibitor for GAL3ST2, i.e. 5′atgtggttcgacttcggct3′ disclosed by Xing-Zhong Wu (Biochemical and Biophysical Research Communications 332 (2005) 934-940), etc.
  • The invention is further set forth by incorporating the detailed Examples. It will be understood that these Examples are only intended to illustrate the invention, rather than restrict the scope of the invention. In the following Examples, the experiments without specified conditions were conducted in accordance with conventional conditions, such as the conditions defined in Sambrook, et al, Molecular Cloning: A Laboratory Guideline. (New York: Cold Spring Harbor Laboratory Press, 1989) or the conditions recommended by the manufacturers. Unless indicated otherwise, percentages and parts were calculated by weight. Unless defined otherwise, all professional terms and scientific phrases used herein have the same meanings as those well known by a person skilled in the art. In addition, any methods and materials similar with or equivalent to the contents recited herein can be applied in the invention. The preferred methods or materials described herein are intended for exemplification only.
  • Example 1 Preparation and Purification of BCMab1 Monoclonal Antibody (1) Preparation of Hybridoma
  • Balb/C mice (commercially purchased from Vital River Laboratory Animal Technology Co. Ltd., Beijing) were immunized with human bladder cancer cell line T24 (ATCC: HTB-4) by intraperitoneally injecting a dose of 1×107 cells/ml PBS. After two weeks, the mice were immunized again with the same injection volume and method. While the serum titer of the mice met the requirement, cell fusion was arranged. The mice were boosted three days before the fusion. At the same time, mouse myeloma cells Sp2/0 (ATCC CRL-1772) were prepared.
  • The sensitized B lymphocytes were fused with the myeloma cells (Practical Immunology, Yang, Y. ed., Changchun Press, published in December, 1994), and selectively cultured (using peritoneal macrophages of the mice as feeder cells) in HAT medium (purchased from Invitrogen Co.; HAT is an enumerate of the initials of hypoxantin, aminopterin and thymidin; HAT medium is thus a cell culture medium containing these three substances).
  • Next, the supernatant of the hybridoma cell culture was detected by ELISA. More specifically, T24 cells (1×105 cells/ml) were coated onto a 96-well plate, 100 μl/well, and cultured at 37° C. overnight. After cell adherence, 4% paraformaldehyde was added, and the plate was fixed at room temperature for min, then washed for 3 times. Subsequently, 100 μl of the supernatant to be detected was added and incubated at 37° C. for 1 hour. Following 3 times of washing, an enzyme-labeled secondary antibody (anti-mouse IgG-HRP) (commercially purchased from Zhongshanjinqiao Biological technology Co. Ltd., Beijing) was added and incubated at 37° C. for 1 hour. Following another 3 times of washing, 50 μl TMB (commercially purchased from Zhongshanjinqiao Biological technology Co. Ltd., Beijing) was added for color development, and kept at room temperature for 5 min. Then 501 stop buffer was added. In the end, the OD values were measured at a wavelength of 450 nm by a microplate reader. An OD value which is higher than that of the negative control by 2 folds or more is considered as a positive result.
  • Then, the selected positive hybridoma cells were clonally cultured (through a limiting dilution, and using peritoneal macrophages of the mice as feeder cells). Following 2-3 rounds of clonal cultures, stable hybridoma cell clones that could generate high-titer monoclonal antibodies were obtained. The hybridoma cell clones were then amplified, cultured, frozen and stored.
  • One type of the positive hybridoma cells in the invention is the hybridoma cell line secreting monoclonal antibodies directed against human bladder cancer. This hybridoma cell line was deposited in China General Microbiological Culture Collection (CGMCC, Beijing, China), with an accession number CGMCC No. 3845, on May 21, 2010.
  • (2) Preparation and Purification of Monoclonal Antibody BCMab1
  • The above hybridoma cells which secrete BCMab1 were inoculated into the abdominal cavities of Balb/C mice, so as to prepare ascites; then the monoclonal antibodies were extracted from the ascites. The monoclonal antibody BCMab1 was purified using Protein G affinity chromatography. First, a Protein G affinity chromatographic column (purchased from “GE” Co.) was prepared, and then balanced with PBS (Phosphate Buffered Saline). The ascites containing monoclonal antibody BCMab1 was passed through the column, then the column was washed with PBS till its OD value approached zero. The column was then eluted with a 0.2 M glycine-HCl solution (pH 2.8), and the eluent was collected. After the OD value of each collecting tube was measured, the eluent with the OD value within the peak range was retained, concentrated by dialysis, then frozen and stored at 20° C.
  • Example 2 Identification of Monoclonal Antibody BCMab1
  • Normal bladder tissue cells HCV29 were purchased from Peking University Health Science Center. Cell lines T24, EJ, HepG2, LoVo, 293, A375, Jurkat, PC-1, MCF-7, K562, Hela, and other cell lines were purchased from ATCC (Rockville, Md., US). Bladder cancer tissues and normal tissues of human were received from The Second Affiliated Hospital of Kunming Medical University, with informed consents. BALB/C mice and nude mice were both purchased from Chinese Academy of Medical Science Institute of laboratory animal.
  • Bladder cancer tissue slices of human (received from Peking University Third Hospital) were stained immunohistochemically with the monoclonal antibody BCMab1 prepared in Example 1 using a conventional method. The result, as shown in FIG. 1, indicated that the bladder cancer tissues of human showed positive reactivity after stained with the monoclonal antibody BCMab1, a secondary antibody (anti-mouse IgG-HRP) and DAB substrate (Zhongshanjinqiao Biological technology Co. Ltd., Beijing); whereas the normal tissues of human showed negative reactivity after being stained with the monoclonal antibody BCMab1, a secondary antibody and substrate.
  • Bladder cancer cells T24 and other cells were detected by the monoclonal antibody BCMab1 using a flow cytometer, and normal human tissues were assayed immunohistochemically. The results, as shown in Table 1 and FIG. 1, indicated that the monoclonal antibody BCMab1 showed a strong positive reactivity with T24 and EJ cells (ATCC CRL-1888), but no cross reactivity with other non-bladder cancer cells or normal tissues of human. Particularly, BCMab was able to stain human bladder cancer cell lines EJ and T24, rather than nonmalignant uretal epithelial cell line HCV29 and systematic tumor cells (FIG. 1 a, 1 b and Table 1). In order to investigate the specificity of BCMab1 to bladder cancer, 123 tumor tissues and 56 normal tissues were screened immunohistochemically. The results indicated that BCMab1 stained bladder cancer tissues only, and showed negative staining for normal bladder tissues and other tissues, such as liver, brain, adrenal gland, pancreas, stomach, colon, breast, lung, ovary, cardiac muscle, thyroid gland, lymph node and marrow (FIG. 1 c, Table 1, unshown data). As a result, BCMab1 can specifically recognize the antigen on membranes of bladder cancer cells.
  • TABLE 1
    Detection of immune responses of monoclonal antibody BCMab1
    directed against human bladder cancer to various cells and tissues
    by flow cytometry and immunohistochemisty
    Cell line BCMab1 Normal tissue BCMab1
    Human bladder cancer cell + Liver
    line EJ
    Human bladder cancer cell + Brain
    line T24
    Normal human uretal epithelial Adrenal gland
    cell line HCV29
    Human hepatic cancer cell line Pancreas
    HepG2
    Human colon cancer LoVo Stomach
    Human malignant melanoma Colon
    cell line A-375
    Human embryonic kidney cell Breast
    line 293
    Human T cell tumor Jurkat Lung
    Human prostate cancer cell Ovary
    line PC-1
    Human breast cancer cell Cardiac muscle
    line MCF-7
    Human cervical cancer cell Thyroid gland
    line HeLa
    Human chronic leukemia cell Lymph node
    line K562
    Human normal peripheral blood Marrow
    monouclear cell
  • Example 3 Identification and Preparation of Antigen AG-α3β1
  • Human bladder cancer cell line T24 was cultured in a RPMI-1640 medium containing 10% fetal bovine serum (purchased from Invitrogen Co.). After digested with 0.25% pancreatin, 1×108 T24 cells were washed with PBS for 3 times. Then 100 μg monoclonal antibody BCMab1 was added, incubated at 4° C. for 2 hours, and washed with PBS for 3 times. The cells were lysed with 1 ml detergent lysis buffer (50 mM Tris-HCl, pH 8.0; 150 mM NaCl; 0.02% sodium azide; 0.1% SDS; 100 μg/ml PMSF; 1 μg/ml Aprotinin; 1% NP-40; 0.5% sodium deoxycholate) for 30 min, and centrifuged at 12,000 g for 10 min. The supernatant was separated and passed through the Protein G affinity chromatographic column. The column was washed with PBS till its OD value approached zero. Subsequently, elution was carried out using a 0.2 M glycine-HCl solution (pH 2.8), and the eluent was collected. The OD value of each collecting tube was measured, and then the eluent with an OD value within the peak range was retained, and identified to be human integrin α3β1 by mass chromatographic analysis (see Table 2).
  • Mass spectrometry: protein gel was cut into small pieces. Pierce Silver Staining Kit and mass chromatographic analysis (Pierce, US) were utilized. Mass chromatographic analysis was conducted for the tryptic peptide generated from digestion of the gel (NanoLC-LTQ, Thermo Finnigan).
  • TABLE 2 
    Mass chromatographic identification of antigen of BCMab I
    Reference Peptide
    Score Coverage MW Accession (Hits)
    Scan(s) Peptide MH+ z XC DeltaCn Sp RSp Ions Count
    integrin alpha 3 isoform b, 10
    precursor [Homo sapiens] 98.30 118756.8 600611 (9 1 0 0 0)
    4184 R.TGAVYLCPLTAHK.D 1431.65517 2 3.44 0.40 817.8 1 16/24 1
    4236 R.YTQVLWSGSEDQR.R 1569.65696 2 4.21 0.44 1330.6 1 20/24 1
    4704 R.YLLLAGAPR.E  974.18150 2 3.14 0.44 1554.8 1 15/16 1
    5265 R.LQSFEGGTVIVIGESGM*K.T 1692.93394 2 3.01 0.19 768.3 1 16/30 1
    5304 K.EAGNPGSLFGYSVALHR.Q 1775.94547 2 3.88 0.57 1213.9 1 21/32 1
    5550 R.HMGAVFLLSQEAGGDLP.R 1802.04787 2 5.98 0.57 3828.0 1 29/32 1
    5600 R.LQSFFGGTVMGESGMK.T 1676.93902 2 4.26 0.49 1569.4 1 22/30 1
    6376 K.LELLLMDNLR.D 1230.50277 2 3.48 0.26 1398.1 1 16/18 1
    6582 R.FAGSESAVFHGFFSMPEMR.C 2135.41062 2 4.61 0.55 1509.1 1 21/36 1
    5265 R.LQSFFGGTVM*GESGMK.T 1692.93394 2 2.45 0.48 581.1 2 15/30 1
    integrin beta 1 ispform 1A 13
    precusor [Homo sapiens] 130.31 88415.9 19743823 (1 3 0 0 0 0)
    3281 K.SAVTIVVNPK.Y 1016.17355 1 1.80 0.29 557.7 2 11/18 1
    3692 R.SNGLICGGNGVCK.C 1336.46409 2 3.03 0.34 820.0 1 18/24 5
    3705 R.DKLPQPVQPDPVSHCK.E 1846.07060 3 3.55 0.38 798.0 1 25/60 5
    3800 K.FCECDNFNCDR.S 1537.55049 2 3.88 0.49 1769.6 1 18/20 5
    4234 K.LSEGVTISYK.S 1097.24343 2 2.62 0.49 972.4 1 15/18 5
    4454 K.WDTGENPIYK.S 1223.31587 2 2.82 0.45 688.0 1 15/18 1
    5306 R.IGFGSFVEK.T  984.13002 2 2.42 0.37 548.6 1 12/16 5
    5792 K.SLGTDLMNEMR.R 1267.45864 2 3.25 0.46 897.1 1 17/20 5
    5979 K.LKPEDITQIQPQQLVLR.L 2020.36187 2 5.29 0.42 1561.2 1 23/32 5
    6170 K.NVLSLTNKGEVFNELVGK.Q 1962.23605 2 6.14 0.55 2084.2 1 24/34 5
    6318 R.LLVFSTDAGFHFAGDGK.L 1782.97638 2 4.78 0.62 1901.6 1 21/32 5
    7665 K.LSENNIQTIFAVTEEFQPVYK.E 2471.74650 2 4.82 0.58 1352.7 1 24/40 5
    8195 R.AEDYPIDLYYLMDLSYSMK.D 2331.64649 2 4.91 0.61 1340.6 1 22/36 5
  • Data were examined by periodate oxidation experiment (Daigo Tsubokawa, Yukinobu Goso, Akira Sawaguchi, et al. A monoclonal antibody, PGM34, against 6-sulfated blood-group H type 2 antigen, on the carbohydrate moiety of mucin. FEBS J. 2007 April; 274(7):1833-48) in combination with antibody-BCMab1 carbohydrate chips (The Consortium for Functional Glycomics, USA). The epitope recognized by antibody BCMab1 is a carbohydrate chain on human integrin α3β1, i.e. [3OSO3]Galβ1-4(Fucα1-3)[6OSO3]GlcNAc (see FIGS. 2, 3 and 4), an O-linked polysaccharide. And such a carbohydrate chain is located on amino acid T (threonine) at position 740 of α3 subunit (on the T in STSS). Since antibody BCMab1 specifically binds to human bladder cancer tissue only, the antigen is indicated to be integrin α3β1 modified by aberrant glycosylation (AG-α3β1), and its epitope [3OSO3]Galβ1-4(Fucα1-3)[6OSO3]GlcNAc is merely expressed in bladder cancer tissue cells. After identification, the structure of [3OSO3]Galβ1-4(Fucα1-3)[6OSO3]GlcNAc is shown below:
  • Figure US20130164216A1-20130627-C00002
  • Detailed procedure of the periodate oxidation experiment are listed as follows: T24 cells were cultured in a 24-well plate for 24 hours. A pre-cooled paraformaldehyde solution (1 ml) was added and kept for 15 min to fix the cells. T24 cells were oxidized by the high-iodine protein in a sodium-acetate balanced solution at room temperature for 1 hour. Excessive periodate was neutralized after cultured with sodium borohydride at room temperature for 30 min. T24 cells were stained immunohistochemically with antibodies BCMab1, anti-MUC1 or anti-CK20. The secondary antibody labeled with peroxidase and DAB assay kit were used for color development.
  • Detailed procedures of the carbohydrate chip experiment for antibody BCMab1 are shown as follows:
  • BCMab1 was diluted with 3% BSA/PBS buffer to a concentration of 1 μg/ml. To a carbohydrate chip, 100 μl BCMab1 diluention was added and incubated in a wet box for 1 hour. The glass slide was rinsed with 0.05% Tween 20/PBS (PBS-T), and incubated with 200 μl Cy5-linked goat anti-mouse IgG antibody (1:200) (Novocastra Laboratories Ltd, UK) in a wet box for 1 hour. The slide was dried in air, scanned to detect fluorescence at a wavelength of 635 nm using a microarray scanner (GenePix 4000B, Molecular Devices), and analyzed with the GenePix Pro software.
  • The experiment for determining the specific position of the polysaccharide epitope recognized by BCMab1 in integrin α3β1 subunits is shown as follows: α3 or β1 subunit was knocked down by RNAi (through pSUPER-shIntegrinα3 or pSUPER-shIntegrinβ1 viral transfection). α3 or β1 subunit-silenced T24 cells and stable cell lines were screened by puromycin (FIG. 5). α3 or β1 could be knocked down by 80%. The α3 or β1 subunit-silenced T24 cells were stained by antibody BCMab1 and analyzed by FACS. The result showed that BCMab1 could not stain the α3-silenced T24 cells (FIG. 6, left), but stain the control cells with blank transfection and the β1-silenced T24 cells (FIG. 6, right). Mouse IgG (mIgG) (Sigma, Germany) was used as a negative control. In sum, the results showed that the site recognized by BCMab1 was in the α3 subunit.
  • The above FACS flow cytometry is shown as follow: first, a single-cell suspension was prepared, and stained on ice for 40 min by adding antibodies; the cells were washed with the PBS containing 0.1% BSA for 3 times, and incubated with FITC-labeled secondary antibody on ice for 40 min; after washing, an analysis was conducted using FACSCalibur flow analysis system (BD, US).
  • The detailed procedures for knocking down integrin α3β1 by RNAi are shown as follows: a small interfering RNA system acting on integrin 3 or β1 was established using pSUPER (Oligoengine, Washington, US) and then cloned into a pSUPER system, so as to express a 19 nt hairpin structure (shRNA) (comprising a small loop of 9 nt). The sequences encoding integrin α3 or β3
  • 5′-GATCCCCGCTACATGATTCAGCGCAATTCAAGAGATTGCGCTG
    AATCATGTAGCTTTTTA-3′ (α3, sense);
    5′-AGCTTAAAAAGCTACATGATTCAGCGCAATCTCTTGAATTGCG
    CTGAATCATGTAGCGGG-3′ (α3, antisence).
    5′-GATCCCCGCTGAAGACTATCCCATTGTTCAAGAGACAATGGG
    ATAGTCTTCAGCTTTTTA-3′ (β1, sense);
    5′-AGCTTAAAAAGCTGAAGACTATCCCATTGTCTCTTGAACAAT
    GGGATAGTCTTCAGCGGG-3′ (β1, antisence).
  • The inserted shRNAs (pSUPER-shIntegrinα3, pSUPER-shIntegrinβ1) were confirmed in these DNA sequences. T24 cells were transfected by Lipofectamin2000 (Invitrogen, US). Integrin α3β1- and GALNT1-knockdown cells were screened out using puromycin. The cells transfected with empty vectors were used as a control.
  • The antigen AG-α3β1 according to the invention can also be prepared by the following standard protocol:
  • I. Preparation of affinity column
    1. Activating 0.33 g CNBr-activated Sepharose 4B Fast Flow matrix with 40 ml of 1 mM HCl for 30 min, and washing it with Binding Buffer 1 (25 mM HEPES, 500 mM NaCl, pH 7.4) twice;
    2. Adding 10 mg antibody BCMab1 for binding at room temperature for 1 h;
    3. Centrifuging, and removing the supernatant; adding 40 ml Blocking Buffer (0.1M ethanolamine, pH 8.0) for blocking at room temperature for 2 h;
    4. Centrifuging, removing the supernatant, loading the column, connecting to a AKTA machine (GE), and rinsing and balancing with Buffer A (25 mM Tris, 150 mM NaCl, pH 7.4), then ready for loading samples.
    II. Cell collection, sample loading and elution
    1. Collecting T24 or EJ cells, and lysing these cells with a lysis buffer (25 mM Tris, 150 mM NaCl, pH 7.4, 1 mM PMSF);
    2. Loading the cell lysate solution, and washing with Buffer A till OD280 does not change significantly;
    3. Eluting the bound protein with Buffer B (25 mM Tris, 2M NaCl, pH 7.4).
    III. Desalinization, purification through molecular sieve, and electrophoretic analysis in combination with mass spectrometric characterization (The first elution peak is AG-α3β1).
  • Example 4 Influence of GAL3ST2 on Aberrant Glycosylation of Integrin α3β1 In Bladder Cancer Tissues
  • In cells, generation of sulphated [3OSO3]Galβ1-4(Fucα1-3)[6OSO3]GlcNAc carbonhydrate required GAL3ST (galactose: 3-O-sulfonyltransferase). GAL3ST, including 4 members i.e. GAL3ST1, GAL3ST2, GAL3ST3 and GAL3ST4, could move sulphates to the position Gal C-3. Surprisingly, compared with other members (GAL3ST1, GAL3ST3 and GAL3ST4), knockdown of GAL3ST2 (galactose: 3-O-sulfonyltransferase 2) could downregulate BCMab1 staining signal to a great extent (FIG. 7). Knockdown of GAL3ST2 could significantly inhibit the growth and migration of cancer. All these results suggested that GAL3ST2 played an important role in the aberrant glycosylation of integrin α3β1 in bladder cancer cells and carcinogenesis.
  • Knockdown of GAL3ST2 was conducted as follows. A small interfering RNA system acting on GAL3ST2 was established using pSUPER and then cloned into a pSUPER system, so as to express a 19 nt hairpin structure (shRNA) (comprising a small loop of 9 nt). The sequences encoding GAL3ST2 shRNA
  • are shown below:
  • 5′-GATCCCCCGGTCACCAACATCATGTTTTCAAGAGAAACATGA
    TGTTGGTGACCGTTTTTA-3′ (GAL3ST2, sense);
    5′-AGCTTAAAAACGGTCACCAACATCATGTTTCTCTTGAAAACA
    TGATGTTGGTGACCGGGG-3′ (GAL3ST2, antisense).
  • The inserted shRNAs (pSUPER-shGAL3ST2) were confirmed in these DNA sequences. T24 cells were transfected by Lipofectamin2000 (Invitrogen, US). GAL3ST2-knockdown cells were screened out using puromycin. The cells transfected with empty vectors were used as a control.
  • The sequences encoding GAL3ST1-, GAL3ST3- and GAL3ST4-knockdown shRNAs are listed below, and the experiments were carried out in the similar procedures with those conducted for the knockdown of GAL3ST2:
  • 5′-GATCCCCTCCGAAACCTGCTCTTCTTTTCAAGAGAAAGAAGA
    GCAGGTTTCGGATTTTTA-3′ (GAL3ST1, sense);
    5′-AGCTTAAAAATCCGAAACCTGCTCTTCTTTCTCTTGAAAAGA
    AGAGCAGGTTTCGGAGGG-3′ (GAL3ST1, antisence).
    5′-GATCCCCGGTGCAGAACATCCTGTTTTTCAAGAGAAAACAGG
    ATGTTCTGCACCTTTTTA-3′ (GAL3ST3, sense);
    5′-AGCTTAAAAAGGTGCAGAACATCCTGTTTTCTCTTGAAAAAC
    AGGATGTTCTGCACCGGG-3′ (GAL3ST3, antisence).
    5′-GATCCCCGGTGCAGAACATCCTGTTTTTCAAGAGAAAACAGG
    ATGTTCTGCACCTTTTTA-3′ (GAL3ST4, sense);
    5′-AGCTTAAAAATCCGCAAGTCACCATCTTTTCTCTTGAAAAAG
    ATGGTGACTTGCGGAGGG-3′ (GAL3ST4, antisence).
  • The detailed procedures for the above cell adhesion experiment are shown as follows:
  • Matrigel (BD, US) was coated on a 96-well plate, and blocked with 10 mg/ml thermally inactivated BSA for 1 hour. After becoming confluence, the above knockdown T24 cells were digested with 1% trypsin, collected at a concentration of 5×105 cell/ml, resuspended in RPMI-1640 containing 1% FBS, and reconstituted at 37° C. for 15 min. shRNA vector was used as a control. The unadhered cells were rinsed off, whereas the adhered cells were collected and stained with 0.1% crystal violet (Sigma, Germany). After decoloration and dye solubilization, the samples were read at 575 nm by ELISA reader.
  • The detailed procedures for the above cell migration experiment are shown as follows:
  • Transwell experiment utilized optimized Boyden chemotaxis kit (with an pore size of 8 μm, Costar, US). Matrigel (100 μg/ml) was coated, and blocked with the PBS containing 1% BSA. The above knockdown T24 cells were digested with 1% trypsin, washed, and resuspended in a medium containing 1% BSA. The lower chambers contained 10% FBS or 1% BSA (negative control). After being incubated at 37° C. for 4 hour, the cells remained in the upper chambers were sucked out, and the cells transferred into the lower chambers were fixed with ethanol and stained with Giemsa. The cells that penetrated through the filter membrane were counted under a microscope. Mitomycin C (2 μg/m) was used to inhibit cell proliferation.
  • Example 5 N-Acetyl-Galactosaminyltransferase 1 (GALNT1) was Highly Expressed in Bladder Cancer Tissues and its Knockdown Line Down-Regulated Integrin α3β1 Glycosylation
  • In order to confirm the function of glycosyltransferase in glycosylation of integrin α3β1, patients suffered from bladder cancer and normal bladder tissues were analyzed by DNA microarray. mRNA samples were obtained from 10 early-staged patients and 10 normal persons through bladder biopsy. It was shown in the microarray analysis that GALNT1 mRNA level was higher in tumor tissues than normal tissues by 11.2 folds (Table 3). In contrast, the levels of other GALNTs (GALNT2-9) and fucosyltransforases (FUT 1-10) did not show significantly difference between bladder cancer tissues and normal tissues (Tables 3 and 4). No significant difference existed in respect of the to expression of α3 and β1 subunit mRNAs either. Housekeeping gene GAPDH was used as a control.
  • The microarray expression analysis is shown as follows detailedly. Total RNA was extracted by isopropanol/chloroform and Trizol from the bladder cancer tissues freshly dissected from 10 cases of patients and 10 cases of normal people. The investigation was carried out using human gene chips containing 35000 genes/chip (Catalog #: 220020; Product Name: CrystalCore human genomewide oligonucleotid microarry chip V2.0; Manufacturer: CapitalBio Corporation). Probes were obtained from 350 μg total RNA of the 10 cases of patients and 10 cases of normal people. cDNA probes were prepared and labeled using CyScribe First-Strand cDNA labeling kit (Amersham Biosciende, US). The bladder cancer tissue specimens were labeled with Cy3; whereas the normal specimens were labeled with Cy5. The labeled cDNA probes were mixed with 2.5×SSC, 0.2% SDS, and 1 μg PolyA RNA. The microarray chips were hybridized at 50° C. for 16 hours, and analyzed by ScanArray 4000 Microarray Analysis System (Packkad Bioscience, US). The data were analyzed by Microarray Analysis Software (Dapple version 0.68 beta).
  • TABLE 3
    High expression of GALNT1 in bladder cancer tissues
    Normal bladder tissues Bladder tumor tissues
    GenBank No. Gene name Signal Detection§ Signal Detection§ Fold-change#
    NM_020474 GALNT 1 303 ± 26 P 3384 ± 142 P 11.2
    NM_004481 GALNT 2 748 ± 23 P 703 ± 35 P 0.9
    NM_004482 GALNT 3 1854 ± 72  P 1315 ± 118 P 0.7
    NM_003774 GALNT 4 36 ± 3 A 42 ± 7 A 1.2
    NM_014568 GALNT 5 12 ± 1 A 16 ± 1 A 1.3
    NM_007210 GALNT 6 815 ± 39 P 902 ± 45 P 1.1
    NM_017423 GALNT 7 735 ± 43 P 836 ± 72 P 1.1
    NM_017417 GALNT 8 87 ± 9 A 107 ± 23 A 1.2
    NM_021808 GALNT 9 172 ± 26 A 166 ± 18 A 0.9
    NM_002204 Integrin alpha-3 3179 ± 112 P 3026 ± 137 P 0.9
    NM_033666 Integrin beta-1 2877 ± 58  P 2800 ± 64  P 1.0
    NM_002046 GAPDH 80942 ± 294  P 81409 ± 367  P 1.0
    Gene expression profile of human normal bladder tissues, and bladder tumor tissues.
    Abbreviations:
    A, absent;
    P, present.
    Calculated mean fluorescence intensity signal.
    §Classification of gene expression as absent or present.
    #Proportion of the signals from experiments with bladder tumor tissues to normal bladder tissues for genes with changed expression (fold change >1 for increased and <1 for decreased gene expression).
  • TABLE 4
    No significant difference of fucosyltransforase (FUT) expression in bladder cancer
    cells and in normal bladder tissues
    Normal bladder tissues Bladder tumor tissues
    GenBank No. Gene name Signal Detection§ Signal Detection§ Fold-change#
    NM_000148 FUT 1 1836 ± 132 P 1684 ± 127 P 0.9
    NM_000511 FUT 2 368 ± 26 P 403 ± 37 P 1.1
    NM_000149 FUT 3 2734 ± 192 P 2835 ± 185 P 1.0
    NM_002033 FUT 4 593 ± 35 P 502 ± 47 P 0.8
    NM_002034 FUT 5 18 ± 3 A 23 ± 5 A 1.3
    NM_000150 FUT 6 1253 ± 29  P 1392 ± 54  P 1.1
    NM_004479 FUT 7 685 ± 73 P 823 ± 67 P 1.2
    NM_178156 FUT 8 406 ± 35 P 378 ± 19 P 0.9
    NM_006581 FUT 9  82 ± 16 A  96 ± 23 A 1.2
    NM_032664 FUT 10 1358 ± 126 P 1407 ± 164 P 1.0
    NM_002046 GAPDH 80942 ± 294  P 81409 ± 367  P 1.0
    Gene expression profile of human normal bladder tissues and bladder tumor tissues.
    Abbreviations:
    A, absent;
    P, present.
    Calculated mean fluorescence intensity signal.
    §Classification of gene expression as absent or present.
    #Proportion of the signals from experiments with bladder tumor tissues to normal bladder tissues for genes with changed expression (fold change >1 for increased and <1 for decreased gene expression).
  • T24 cells were transfected with pSUPER-shGALNT1 vectors, so as to downregulate GALNT1 expression. The GALNT1-knockdown T24 cell lines that had been transfected stably were screened out using puromycin (FIG. 8). GALNT could be silenced by 80%. Compared with empty pSUPER vector, the GALNT1-knockdown cell lines could downregulate BCMab1 staining signal significantly (FIG. 9, left). A competitive inhibition experiment was conducted to the substrate BG of GALNT1. In the BG experiment, signal of integrin α3 μl recognized by BCMab1 was downregulated (FIG. 9, right). mIgG was the negative control. The result showed that GALNT1 might induce aberrant methylation of the integrin α3β1 in bladder cancer.
  • The method for silencing GALNT1 is shown below. A small interfering RNA system acting on GALNT1 was established using pSUPER and then cloned into a pSUPER system, so as to construct a pSUPER-shGALNT1 vector and express a 19 nt hairpin structure (shRNA) (comprising a small loop of 9 nt). The sequences encoding GALNT1 shRNA are shown below:
  • 5′-GATCCCCCCAGTACAAAAGCCTCATGTTCAAGAGACATGAGG
    CTTTTGTACTGGTTTTTA-3′ (GALNT1, sense);
    5′-AGCTTAAAAACCAGTACAAAAGCCTCATGTCTCTTGAACATG
    AGGCTTTTGTACTGGGGG-3′ (GALNT1, antisence).
  • The inserted shRNAs (pSUPER-shGALNT1) were confirmed in these DNA sequences. T24 cells were transfected by Lipofectamin2000 (Invitrogen, US). GALNT1-knockdown cells were screened out using puromycin. The cells transfected with empty vectors were used as a control.
  • GALNT1 was inhibited by BG. T24 cells were cultured in a 6-well plate with or without 2 mM BG (Sigma, Germany) for 7 days. The cells were collected, into which BCMab1 and its corresponding FITC-linked secondary antibody were added and incubated for 40 min. The expression of antigen of BCMab1 was analyzed by FACS.
  • Example 6 Knockdown of Integrin α3 or GALNT1 can Inhibit Cancer Proliferation and Tumor Growth
  • Knockdown of integrin α3 or GALNT1 (see above for the detailed procedures) could significantly reduce the binding rate between [3H]-thymidine and DNA of T24 cells (FIG. 10, a), and remarkably affect the cloning of T24 cells (FIG. 10, b). In order to investigate the influence of the knockdown of integrin α3 or GALNT1 on tumors in vivo, a tumor-bearing model was constructed by inoculating BALB/c nude mice at their backs subcutaneously, then tumor change was observed by injecting vectors. Compared with the control group, the integrin α3- or GALNT1-knockdown group significantly inhibited growth of the subcutaneous tumors (FIG. 10, c). And by the end of the observation period, i.e. on Day 35, the tumor inhibition ratio reached 63% and 42%, respectively.
  • The steps of the cell proliferation experiment are shown as follows. 3H nuclide incorporation assay was utilized. Wildtype T24 cells and shIntegrin α3- or shGALNT1-knockdown T24 cells (2×103 cells/well) were coated onto a 96-well plate and cultured till reaching 70%-80% confluence. The cells were cultured in serum-free RPMI-1640 for 24 hours and the medium was replaced by RPMI-1640 containing 10% FBS. To the wildtype T24 cells, 10 μg/ml of BCMab1 or control mIgG was added. After 72 hours, 3H was added and the cells were cultured for 4 hours. Then the cells were collected and detected for 3H incorporation amount by a liquid scintillation counter LKB1219.
  • The above animal experiment was shown below. BALB/c nude mice (female, 6 weeks old, with a body weight of about 15 g) (purchased from Chinese Academy of Medical Science Institute of laboratory animal) were fed in a SPF environment. T24 cells (including wildtype T24 cells and shIntegrin α3- or shGALNT1-knockdown T24 cells) were xenografted, and tumor cells (1×107, suspended in PBS) were injected subcutaneously at their backs. When tumors grew up to 3-5 mm, the mice with wildtype T24 were grouped (10 per group), and BCMab1 or mIgG (10 mg/kg) was injected intraperitoneally, 3 times/week, for 35 days. The tumor volumes were measured twice per week. Orthotopic bladder cancer transplantation model, T24 cells were labeled with GFP, and tumors were induced to be transplanted into bladders by blood vessel probes. BCMab1 or mIgG (1 mg/kg) was administered into the bladders of 24 mice in each group. The images were obtained by an in vivo imaging system (IVIS) on days 5, 10, 15, 20, 25 and 30 after transplantation.
  • Example 7 Knockdown of Integrin α3 or GALNT1 can Inhibit Cancer Proliferation and Tumor Growth
  • The detailed procedures for knockdown of integrin α3 or GALNT1 are described in the previous Examples. The function of the integrin α3- or GALNT1-knockdown T24 cells was detected. It was found that α3-knockdown T24 cells could significantly inhibit adhesion between cells and Matrigel (FIG. 11, a). The migration experiment indicated that α3-knockdown T24 cells significantly inhibited cell migration (FIG. 11, b). The scratch test (Huang, J., et al. Genetic and epigenetic silencing of SCARAS may contribute to human hepatocellular carcinoma by activating FAK signaling. J Clin Invest 120, 223-241 (2010)) showed similar results (FIG. 11, c). However, knockdown of GALNT1 did not impact cell adhesion or migration significantly (FIG. 11, a-c), suggesting that silence of integrin α3 could inhibit adhesion and migration of bladder cancer cells.
  • Example 8 Knockdown of Integrin α3 or GALNT1 can Enhance NK Cell-Mediated Lysis of Bladder Cancer Cells
  • The detailed procedures for knockdown of integrin α3 or GALNT1 are described in the previous Examples. We investigated the killing action of nature killer (NK) cells against integrin α3- or GALNT1-knockdown T24 cell lines. Compared with the empty vector controls (Vector/integrin α3 and Vector/GALNT1), the knockdown cell lines could enhance the killing action of NK cells (extracted from peripheral blood of healthy human). In the experiment for testing the killing action of NK cells against α3-knockdown cell lines, where E/T (effector cell/target cell) ratios were 10:1, 20:1, and 40:1, the killing activities of NK cells against α3 knockdown and empty vector controls were 12.5±1.6% vs 10.8±0.6% (p=0.106), 24.6±3.9% vs 18.5±2.1% (p=0.001), 1 to and 43.8±5.4% vs 30.7±2.7%, respectively (p<0.001) (FIG. 11, d, left). In the experiment for testing the killing action of NK cells against GALNT1-knockdown cell lines, the killing activities were 12.4±0.8% vs 11.7±1.4% (p=0.439), 20.5±2.1% vs 17.3±2.3% (p=0.017), and 33.2±4.4% vs 27.6±3.7% (p<0.002), compared with controls. In conclusion, knockdown of α3 could activate NK cell-mediated killing action more effectively, compared with knockdown of GALNT1. And similar results were achieved for NK92 cell (ATCC: CRL-2407) mediated killing action (FIG. 11, d, right).
  • To further study the killing action of NK cells against tumor cells, the NK cells were eliminated by anti-asialo GM1 antibody (Wako Chemicals, Richmond, Va.) in BALB/C mice before inoculation of T24 cells. In this model, integrin α3- or GALNT1-knockdown T24 cells could significantly inhibit growth of tumors, compared with empty vectors. No died cell was found in the tumor tissues of α3- or GALNT1-T24-inoculated mice from which NK cells had been eliminated (FIG. 11, f); whereas a few died cells were observed in the early-stage tumor tissues from which NK cells had not been eliminated. Interestingly, in the tumors of the mice inoculated with α3-/GALNT1-T24, NK cells could significantly facilitate the expression of perforin and IFNγ (FIG. 11, g, h). Moreover, the number of the NK cells was greater in shIntegrin α3 or shGALNT1-treated tumors than in the control group (FIG. 11, i). The data suggested that the killing action mediated by NK cells played an important role in the tumor-bearing mouse with α3- or GALNT1-knockdown T24.
  • Example 9 Antibody BCMab1 Inhibited Proliferation of Tumor Cells
  • T24 cells were inoculated into a 96-well plate in a density of 1×103 cells/well, and cultured conventionally till reaching the confluence of 70-80%. Then the cells were starved in a serum-free medium for 24 hours. Antibody BCMab1 (100 μg/mL) was added, and the culture was continued for 48 hours while serum was resumed. At the same time, homogenic mouse IgG (mIgG (Sigma, Germany)) was used as a control. The cells were incubated with the incorporation of [3H]-TdR (synthesized by Isotope Institute of China Institute of Atomic Energy) for 4 hours, and counted for the amount labeled by [3H]-TdR using a liquid scintillation counter LKB1219 (LKB Corporation, Sweden). As shown in FIG. 12, the result indicated that compared with the control group, the proliferation of tumor cells was inhibited significantly in the antibody BCMab1-treated group.
  • Example 10 BCMab1 and BCMab1-Ra Inhibited Growth of Subcutaneous and Orthotopic Bladder Cancer Models
  • As shown in FIGS. 10 and 11, BCMab1 could significantly inhibit the growth of bladder tumors, colony formation and migration. For the sake of confirming the anti-tumor function of BCMab1 in vivo, a xenograft tumor model was established by injecting T24 cells (1×107, suspended in PBS) into the backs of BALB/c nude mice. When tumors grew up to a diameter of 0.3-0.5 cm, BCMab1 or control mIgG antibody (10 mg/kg) was injected intraperitoneally, 3 times/week. Compared with the control, tumors were reduced significantly in the BCMab1-treated group (FIG. 12, a). And the inhibition ratio reached 77% by the end of the experiment (Day 40).
  • To detect the orthotopic anti-tumor function of BCMab1, orthotopic transplantation tumor model of bladder cancer was built up using GFP fluorescence labeled T24 cells (GFP-T24). GFP-T24 (5×106) cells were transplanted into bladder. GFP was detected by an in vivo imaging system (IVIS), so as to confirm that all the inoculated BALB/c nude mice were suffered from bladder cancer. The mice were grouped (24/group). BCMab1 or mIgG 1 mg/kg was injected into bladders (FIG. 12, b). Compared with the control group, administration of BCMab1 in bladder effectively inhibited growth of bladder cancer and extended lift time of T24 tumor-bearing mice (p<0.0001) (FIG. 12, c, d). No BCMab1-related toxicity was found in the experiment. And no metastasis was found in the BCMab1-treated group by histopathological investigation. However, in the mIgG control group, tumor metastasis was present in 30%-50% mice, tumors metastasized to lungs and lymph nodes, and 10% mice died (data not shown).
  • Immunotoxin BCMab1-Ra was generated through the conjugation between BCMab1 and ricin A chain. More specifically, ricin A chain and BCMab1 were connected via a heterogeneous difunctional crosslinking agent SPDP (N-succinimidyl 3-(2-pyridyldithiol)propionate) into the immunotoxin BCMab1-Ra. The detailed procedure was shown as follows.
  • 1. Reaction (RT-PDP) Between Ricin A Chain (Ra) and Crosslinking Agent (SPDP):
  • (1) 10 mg/3.5 ml Ra (prepared in accordance with: Song, P et al., The preparation of conjugate composed of ricin A chain and mAB 3G11 and the evaluation of its biological activity in vitro, Journal of Chinese Hospital Pharmacy, Vol. 25, No. 10, p899-901, 2005) was added to 100 μl 4 mg/ml SPDP (Sigma-Aldrich Co. LLC.) (dissolved in absolute ethanol, formulated immediately before use) in a 10-fold excessive molar ratio, and agitated at room temperature for 30 min;
    (2) A small amount of precipitate was observed after agitation, and then centrifuged at 10000 rpm for 30 seconds;
    (3) The supernatant was collected and placed into a dialysis bag (Beijing Xin Jing Ke Biotechnology Co. Ltd (MD44120)), then dialyzed with acetate buffer (0.02M/pH 4.5) overnight;
    (4) A small amount of precipitate was observed during the dialysis, and removed by centrifugation; the remaining solution was Ra-PDP solution.
  • 2. Reaction (Ra-SH) Between Ra-PDP and DTT
  • (1) 27 mg DTT (Sigma-Aldrich Co. LLC.) (with a final concentration of 50 mM) was added to 3.5 ml Ra-PDP, and agitated at room temperature for 30 min;
    (2) A small amount of precipitate was observed after agitation, and then centrifuged at 10000 rpm for 30 seconds;
    (3) The supernatant was collected and placed into a dialysis bag (Beijing Xin Jing Ke Biotechnology Co. Ltd (MD44120)), then dialyzed with PBS (containing 0.15M NaCl, 0.02M/pH 7.4) repeatedly till smellless.
  • 3. Reaction (BCMab1-PDP) Between Antibody (BCMab1) and Crosslinking Agent (SPDP):
  • (1) 5 mg/6.5 ml antibody was added to 30 μl of 4 mg/ml SPDP (dissolved in absolute ethanol, formulated immediately before use) in a 10-fold excessive molar ratio, and agitated at room temperature for 30 min;
    (2) A small amount of precipitate was observed after agitation, and then centrifuged at 10000 rpm for 30 seconds;
    (3) The supernatant was collected and placed into a dialysis bag, then dialyzed with PBS (containing 0.15M NaCl, 0.02M/pH 7.4).
  • 4. Conjugation of Immunotoxin (BCMab1-Ra):
  • (1) 3.5 ml Ra-SH and 6.5 ml BCMab1-PDP were mixed (10 ml), and agitated at room temperature overnight (more than 16 hours);
    (2) Obvious turbidity was observed, and centrifuged at 10000 rpm for 1 min;
    (3) The samples were concentrated by PEG20000 into a loading volume of 5 ml and loaded onto a G-100 column (Beijing Xin Jing Ke Biotechnology Co. Ltd (3098));
    (4) Each eluting peak was collected after loading samples (eluant: PBS (containing 0.15M NaCl, 0.02M/pH 7.4)), wherein the first peak was immunotoxin BCMab1-Ra, the second peak was antibody, and the third peak was toxin; measurement results showed immunotoxin OD280=0.116, i.e. 5.1 mg/22 ml;
    (5) immunotoxin was filtered for sterilization, and then kept at 4° C. or frozen/stored at −20° C.
  • The detailed steps are shown as follows:
  • Figure US20130164216A1-20130627-C00003
  • It was shown in indirect immunofluorescence, FACS analysis and competitive binding experiments that the binding activity between the antibody and immunotoxin was 95% of the initial activity. The cell culture suggested that BCMab1-Ra was a very effective biological agent capable of killing bladder cancer cell lines (EJ and T24). BCMab1-Ra could inhibit the proliferation and growth of human bladder cancer cell lines in vitro (data not shown). Interestingly, direct infusion of BCMab1-Ra into bladders through urethral injection could inhibit tumor growth in mouse orthotopic transplantation tumor 1 to model which had the same survival rate as normal mice (FIG. 12, b-d). No toxicity caused by BCMab1-Ra was observed. As a result, BCMab1-Ra can be used as a potential toxic medicament for the treatment of bladder cancer.
  • Twenty-five sites were detected in the bladder of a mouse, so as to determine whether bladder cancer was induced. Through IVIS detection of GFP fluorescence, it was found that all mice were suffered from bladder cancer. On Day 25 during the treatment, all tumors disappeared in the BCMab1 group (FIG. 12, e, middle). The immunohistochemical and HE staining results showed that only a few cancer cells remained at the inoculation sites of T24 cells; and no tumor cell was detected in the BCMab1-Ra-treated group immunohistochemically (data not shown). However, the tumors did not disappear in the mIgG control group (FIG. 12, e, left). No appreciable damage was observed on the bladder mucous membranes of the mice in both BCMab1 and BCMab1-Ra groups (FIG. 12, e). Neither of them showed any difference in the bladder mucous membranes from normal mice under microscope. Overall, BCMab1 could inhibit growth of bladder cancer in vivo, and a part of mice, without any severe side effect, were healed completely. In our experiments, BCMab1-Ra also could eliminate bladder cancer cells without causing side effect.
  • In order to further investigate whether BCMab1 impacts other cells in a microenvironment of tumor, we detected microvessel density (MVD) and cytokines such as VEGF and IL-6 in a tumor-bearing mouse model. In the BCMab1 treatment group, the number of MVD was reduced significantly (FIG. 12, f), generation of VEGF and IL-6 was inhibited in tumors (FIG. 12, g, h). Same results were shown in the BCMab1-Ra treatment group as well (data not shown). It was suggested that BCMab1 could also suppress angiogenesis while inhibiting proliferation of tumors.
  • Furthermore, we conducted some researches on the anti-tumor molecular mechanism of BCMab1, and found that BCMab1 could cause stasis of tumor cell cycles by preventing focal adhesin kinase (FAK) and c-Jun from phosphorylation (FIG. 16, a, b). The glycosylated α3β1 on T24 cells enhanced FAK-dependent signalings (FIG. 16, c). Knockdown of integrin α3 or GALNT1 lowered the phosphorylation levels of FAK and c-Jun significantly. In the case that NK cells were removed, BCMab1 still could inhibit growth of cancer (FIG. 16, d). All these results suggested that BCMab1 could inhibit tumor growth and improve the killing function of NK cells.
  • The above signal pathway experiment is shown as follows. T24 cells were starved in serum-free RPMI-1640 for 24 hours, and then cultured in RPMI-1640 containing 10% FBS and 1 μg/ml BCMab1 or mIgG control. After 48 hours, the cells were lysed with lysis buffer. The lysate supernatant was analyzed by immunodotting with anti-human FAK, FAK, Phospho-FAK (Tyr397), c-Jun, Phospho-c-Jun (Ser63), Phospho-c-Jun (Ser73), CyclinD1 or CDK4 antibodies.
  • The antibodies used in the above experiments include: rabbit anti-human CyclinD1, rabbit anti-human CDK4, rabbit anti-human c-Jun, Phospho-c-Jun (Ser63), Phospho-c-Jun (Ser73), rabbit anti-human FAK, Phospho-FAK (Tyr397) (CST, US), and homotypical control mIgG (Sigma, Germany). Mouse monoclonal antibody BCMab1 was isolated from ascites using Protein A Sepharose. The biotinylatd HRP and FITC labeled secondary antibodies were purchased from Pierce, US and Sigma, Germany, respectively.
  • Integrins impacted the adhesion-related effects in tumor cells, including proliferation, survival, migration, invasion and passage of FAK signal pathway. Phosphorylation of FAK was deemed as one of the major steps realizing further phosphorylation of Src and Fyn and triggering of signal pathway. Glycosylation of integrin α3β1 was clearly different in tumor cell lines and in the control cell line. The connection between cells and extracellular matrix protein was modulated by glycosylation. We found that knockdown of integrin α3 or GALNT1 could inhibit proliferation of cancer and growth of tumors in vivo and in vitro. Suppression of integrin α3β1 signaling by BCMab1 also could inhibit proliferation of cancer and growth of tumors in vivo and in vitro. BCMab1 treatment downregulated phosphorylation of FAK and c-Jun, reduced expression level of cyclin D1 and CDK4, and interfered with cell proliferation and cell cycles. Therefore, aberrantly glycosylated integrin α3β1 might result in malignant transformation of cells. BCMab1 could suppress tumor growth by inhibiting angiogenesis in the microenvironment of bladder cancer tumor. BCMab1 downregulated the generation of VEGF and IL-6 in tumors, which suggested that targeting of tumor microenvironment required further investigation.
  • Example 11 Consistency Between Aberrant Glycosylation Level of Integrin α3β1 and the Severity Degree and Lifetime of Cancer Patients
  • In order to determine whether the aberrant glycosylation expression level of integrin α3β1 is consistent with the clinical manifestation of cancer patients, immunohistochemical assays were conducted to the patients suffering from bladder cancer. The bladder cancer tissues obtained from 69 patients in Grades 1-3 (G1-3) and Stages 1-4 (T1-4) were detected. Weak staining occurred in the patients with low grades (G1-G2) and low stages (T1-T2) (FIG. 17, a and Table 5); whereas intense BCMab1 staining occurred in the patients with a high grade and high stages (G3 and T3, T4). The staining of normal bladder tissue was used as a control. As a result, the aberrant glycosylation expression level of integrin α3β1 was consistent with the severity degrees of cancer patients.
  • We also analyzed the aberrant glycosylation expression of integrin α3β1 and 1 to clinical pathological features in the patients who had experienced radical cystectomy. The aberrant glycosylation expression of integrin α3β1 was higher in the bladder cancer patients with high stages, deep invasion and lymph-node involvement than in the patients with low grades, shallow invasion and no lymph-node involvement (Table 5). The patients with a higher aberrant glycosylation expression of integrin α3β1 had a lower survival rate than the patients with lower expression (P>0.0036) (FIG. 17, b).
  • TABLE 5
    Integrin α3β1 modified by abnormal glycosylation is intimately
    correlated with clinical pathology of bladder cancer tissues
    Expression of
    aberrantly glycosylated
    Integrin α3β1
    Total Patients High Low P
    Age Mean 62.3 65.7 58.9 0.352
    Sex Male 48 25 23 0.732
    Female 21 10 11
    Tumor stage Ta, Tis, T1 26 5 21 <0.001
    T2 15 9 6
    T3 20 13 7
    T4 8 5 3
    Grade G1 or 2 22 5 17 <0.001
    G3 47 32 15
    Configuration Papillary 17 6 11 0.108
    Nonpapillary 52 30 22
    Number of tumors Solitary 38 23 15 0.071
    Multiple 31 12 19
    Lymphatic invasion Negative 18 2 16 <0.005
    Positive 39 21 18
    Unknown 12 7 5
    Follow-up (month) Mean 72.6 70.3 74.9 0.478
  • Bladder cancer is a common disease with an incidence increasing year after year. More and more evidence demonstrated that molecular signal pathway affected cell stability and thus caused bladder cancer. Cell growth mainly involves five aspects, including cell cycle, cell death, cell growth, signal transduction, and gene regulation. Both the influence of external carcinogenic signals or variation of genetic factors may result in signal disorder. Maintenance and progress of tumors depend on two external factors, i.e. the correlations between interstitial substance and knizocyte, and between angiogenesis and tumor cell invasion. However, the molecular mechanism for malignant transformation of bladder cancer was still unknown. The novel monoclonal antibody BCMab1 prepared by the inventors could specifically bind to aberrantly glycosylated integrin α3β1 and recognize aberrantly glycosylated α3 subunit. High expression of glycosyltransferase GALNT1 may result in aberrant glycosylation of integrin α3. Knockdown of integrin α3 or GALNT1 could inhibit proliferation of cancers and growth of tumors in vivo and in vitro. As such, block of integrin α3 signal by BCMab1 could also inhibit proliferation of cancers and growth of tumors. In addition, aberrant glycosylation level of integrin α3β1 was correlated with clinical staging and prognosis of bladder cancer. The results suggested that aberrant glycosylation of integrin α3β1 was correlated with occurrence of bladder cancer. Aberrant glycosylation of integrin α3β1 could become a new diagnosis marker for bladder cancer and a new indicator for prognosis.
  • Example 12 Preparation of Immunodiagnosis Reagent for Bladder Cancer
  • Using the antigen AG-α3β1 and antibody BCMab1 described in the invention, a hypersensitized competitive ELISA method is developed in the invention to detect bladder tumors in human urine. The major process includes steps of using antigen AG-α3β1 as an immobile phase and standard sample; using antibody BCMab1 as a detection antibody in combination with the secondary antibody (horseradish peroxidase (HRP) labeled goat-anti-mouse IgG); and detecting bladder cancer antigen AG-α3β1 in urine of normal human and bladder cancer patients. It was proved by the experiments that the immunodiagnosis reagent for bladder cancer according to the invention had, compared with prior art, the following positive effects: (1) it directly detects urine liquid without injury, so as to avoid inconvenience and distress caused to patients by cystoscopy; (2) it has a high sensitivity and specificity that are much higher than those of the detection of current bladder tumor marker (such as nuclear matrix protein, cytokeratin, hyaluronic acid and the like) and pathological examination of urine exfoliative cells; (3) it has a high specificity and sensitivity and utilizes competitive ELISA to detect bladder cancer antigen in cells, so as to avoid the defects of higher factitiousness and lower sensitivity which cannot be overcome by cell pathology itself; and (4) it is convenient and fast, thereby suitable for screening early-stage bladder cancer patients and detecting relapse of tumors.
  • Detailed experiment procedure is shown as follows:
  • 1) Sample source and clinical data: urine of healthy volunteers and bladder cancer patients were obtained from Peking University Third Hospital.
    2) Detailed steps:
      • i) coating ELISA plate with antigen: antigen AG-α3β1 was diluted to 1 μg/mL with PBS and coated on a plate, 5 μg/well, at 4° C. overnight.
      • ii) blocking nonspecific binding sites: incubated with 2% BSA/PBS, 200 μg/well, at 37° C. for 2 hours.
      • iii) incubating samples: the samples to be tested were fresh urine; the loading amount was 50 μL; urine obtained from healthy volunteers was used as the negative control; 1 μg/mL antibody BCMab1 (50 μL) was added to each well; each sample was duplicated and incubated at 37° C. for 1 hour.
      • iv) washing with PBST for 5 times.
      • v) adding horseradish peroxidase (HRP) labeled goat-anti-mouse IgG antibody (Sigma Co.), 10 μL/well, incubating at 37° C. for 1 hour.
      • vi) washing with PBST for 5 times.
      • vii) adding substrate (tetramethyl benzidine, H2O2), 100 μl/well, incubating in dark at room temperature for 10-20 min; adding 2M H2SO4 (50 μL) to terminate the reaction; measuring OD (450 nm) value by an Microplate reader.
        2) Result analysis: according to negative OD value/OD critical value≈2.1, OD critical value≈negative OD value/2.1; then the sensitivity and specificity 1 to could be determined in accordance with OD critical value.
  • Actual Samples
    + Total
    Detection + 82 7 89
    Results 18 93 111
    Total 100 100 200
  • Clinical sensitivity can be used to evaluate the capability of a certain assay to detect sick subjects, and sensitivity is the proportion of true positives correctly identified in the actual sick subjects.
  • In the experiment, sensitivity=82/(82+18)×%=82%.
  • Clinical specificity can be used to evaluate the capability of a certain assay to identify healthy subjects, and specificity is the proportion of true negatives correctly identified in the actual healthy subjects.
  • In the experiment, specificity=93/(7+93)×%=93%.
  • Example 13 Screening for Potential Medicaments to Treat Bladder Cancer
  • T24 cells, endogenously expressing GAL3ST2 protein, were taken and cultured as above. The cells were used as cell models to screen for the medicaments to treat bladder cancer in mammals.
  • Test group: the above cell culture was treated with a candidate;
    Control group: the above cell culture was not treated with the candidate.
  • After treated for a proper time period, the cells were detected for GAL3ST2 expression, enzyme activities, existing amount or secretion status by 1 to conventional methods. Compared with the control group, GAL3ST2 protein expression, enzyme activities, existing amount or secretion was significantly reduced by 30% or more in the test group, which indicated that such a candidate is a potential substance to treat bladder cancer.
  • Confirmation: RNAi sequence of GAL3ST2 was tested as a candidate. The result showed that GAL3ST2 protein expression was inhibited. Thus the RNAi sequence is a potential substance to treat bladder cancer.
  • Example 14 Screening for Potential Medicaments to Treat Bladder Cancer
  • T24 cells, endogenously expressing N-acetylgalactosaminyltransferase 1 protein, were taken and cultured as above. The cells were used as cell models to screen for the medicaments to treat bladder cancer in mammals.
  • Test group: the above cell culture was treated with a candidate;
    Control group: the above cell culture was not treated with the candidate.
  • After treated for a proper time period, the cells were detected for N-acetylgalactosaminyltransferase 1 expression, enzyme activities, existing amount or secretion status by conventional methods. Compared with the control group, N-acetylgalactosaminyltransferase 1 protein expression, enzyme activities, existing amount or secretion was significantly reduced by 30% or more in the test group, which indicated that such a candidate is a potential substance to treat bladder cancer.
  • Confirmation: RNAi sequence of N-acetylgalactosaminyltransferase 1 was tested as a candidate. The result showed that N-acetylgalactosaminyltransferase 1 protein expression was inhibited. Thus the RNAi sequence is a potential substance to treat bladder cancer.
  • Although the detailed Examples of the invention are illustrated as above, it can be understood that the invention can be conducted in a practice other than the above illustrations, and the protection scope of the invention is not limited by the description.

Claims (18)

1. An aberrantly glycosylated integrin AG-α3β1 characterized in that the integrin α3β1 has a carbohydrate structure [3OSO3]Galβ1-4(Fucα1-3)[6OSO3]GlcNAc as an antigenic epitope,
wherein the carbohydrate structure [3OSO3]Galβ1-4(Fucα1-3)[6OSO3]GlcNAc is represented by a structural formula shown as follow:
Figure US20130164216A1-20130627-C00004
2. The AG-α3β1 according to claim 1, wherein the integrin α3β1 has an α3 subunit having an amino acid sequence shown as SEQ ID No: 1 and a β1 subunit having an amino acid sequence shown as SEQ ID No: 2; preferably the aberrant glycosylation is located on the α3 subunit, and more preferably on the amino acid T (threonine) at position 740 of the α3 subunit.
3. A binding molecule directed against the AG-α3β1 according to claim 1, wherein the binding molecule can specifically recognize or bind to the carbohydrate structure [3OSO3]Galβ1-4(Fucα1-3)[6OSO3]GlcNAc as the antigenic epitope; the binding molecule is a polyclonal antibody or monoclonal antibody.
4. The binding molecule according to claim 3, which is an anti-AG-α3β1 monoclonal antibody BCMab1, and the monoclonal antibody is secreted by the hybridoma cell line deposited as CGMCC No. 3845.
5. The binding molecule according to claim 3, wherein the binding molecule is conjugated with a substance selected from the group consisting of a biological marker, an antitumor drug, a toxin and a radioactive agent.
6. The binding molecule according to claim 3, wherein the biding molecule is formulated in a pharmaceutical composition, that comprises an effective dose of the binding molecule and a pharmaceutically acceptable carrier, diluents or excipient, and wherein the binding molecule is optionally conjugated with a substance selected from the group consisting of a biological marker, an antitumor drug, a toxin, and a radioactive agent.
7. A kit for detecting bladder cancer, comprising the binding molecule according to claim 3 or AG-α3β1; and optionally another reagent for detecting bladder cancer, wherein the binding molecule is optionally conjugated with a substance selected from the group consisting of a biological marker, an antitumor drug, a toxin, and a radioactive agent.
8. The kit according to claim 7, wherein the detection is carried out by an enzyme linked immunosorbent assay.
9. The kit according to claim 7, wherein the sample to be tested is urine or bladder tissue.
10. A hybridoma cell line secreting antibody BCMab1 directed against integrin α3β1 of human bladder cancer, wherein the hybridoma cell line is deposited as CGMCC No. 3845.
11. A method for treating bladder cancer, comprising administering to a subject in need thereof an effective amount of the pharmaceutical composition according to claim 6.
12. The method according to claim 11, wherein the pharmaceutical composition is used in a combination therapy with another drug for the treatment of bladder cancer; said another drug for the treatment of bladder cancer is selected from the group consisting of antitumor chemotherapeutic agent, immunoreinforcing agent, and cytokines; wherein said pharmaceutical composition and said another drug for the treatment of bladder cancer are used simultaneously, separately or sequentially for the treatment of human or animal bodies.
13. A method for diagnosing or prognosing bladder cancer using the AG-α3β1 according to claim 1 or an antibody against the AG-α3β1, the method comprising:
reacting the antibody with a test sample to determine an amount of antigen-antibody binding; or
adding the AG-α3β1 to a reaction mixture containing a test sample and the antibody to determine an extent of reduction of antigen-antibody binding by the added AG-α3β1.
14. A method, using the AG-α3β1 according to claim 1, for preparing or screening antibodies, comprising:
injecting the AG-α3β1 as an immunogen to immunize an animal, so as to obtain a monoclonal or polyclonal antibody; or
applying the AG-α3β1 as an antigen to screen and prepare antibodies by a phage, yeast or ribosome display technique.
15. A method for treating bladder cancer, comprising administering to a subject in need thereof an RNAi sequence directed against GAL3ST2.
16. A method for treating bladder cancer, comprising administering to a subject in need thereof an inhibitor of N-acetylgalactosaminyltransferase 1, wherein the inhibitor is selected from the nucleotide sequence of the RNAi as shown by SEQ ID No: 4, uridine 5′-phosphoric (1-hexadecanesulfonic) anhydride, and Ammonium (3-Acetamido-2,6-anhydro-3-deoxy-D-glycero-L-gluco-heptitol-1-yl)(Uridine-5′-yl)Diphosphate.
17. A process of screening for a potential substance to treat bladder cancer, comprising the steps of:
(1) treating a system expressing N-acetylgalactosaminyltransferase 1 protein or GAL3ST2 protein with a candidate; and
(2) detecting the expression or activity of the N-acetylgalactosaminyltransferase 1 protein or GAL3ST2 protein in the system;
wherein, if the candidate can reduce the expression or activity of the N-acetylgalactosaminyltransferase 1 protein or GAL3ST2 protein, then the candidate is indicated to be a potential substance to treat bladder cancer.
18. A method for diagnosing or prognosing bladder cancer, comprising determining in a test sample an activity of N-acetylgalactosaminyltransferase 1.
US13/814,446 2010-08-11 2011-08-12 Bladder cancer tumor marker, antibody and use thereof Active 2032-01-08 US9073995B2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN201010251384.6 2010-08-11
CN2010102513846A CN102372773B (en) 2010-08-11 2010-08-11 Human bladder cancer tumor marker, antibody thereof and application thereof
CN201010251384 2010-08-11
PCT/CN2011/001345 WO2012019437A1 (en) 2010-08-11 2011-08-12 Bladder cancer tumor marker, antibiotic and application thereof

Publications (2)

Publication Number Publication Date
US20130164216A1 true US20130164216A1 (en) 2013-06-27
US9073995B2 US9073995B2 (en) 2015-07-07

Family

ID=45567321

Family Applications (1)

Application Number Title Priority Date Filing Date
US13/814,446 Active 2032-01-08 US9073995B2 (en) 2010-08-11 2011-08-12 Bladder cancer tumor marker, antibody and use thereof

Country Status (3)

Country Link
US (1) US9073995B2 (en)
CN (1) CN102372773B (en)
WO (1) WO2012019437A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2019513410A (en) * 2016-02-19 2019-05-30 ビーアンドエイチ バイオテクノロジーズ,エルエルシーB&H Biotechnologies, Llc Anti-N-acetylglucosamine and N-acetylgalactosamine antibodies
WO2020257296A1 (en) * 2019-06-21 2020-12-24 Board Of Regents, The University Of Texas System Targeting alpha3beta1 integrin for treatment of cancer and other diseases
US11578134B2 (en) * 2017-08-14 2023-02-14 Chong Li Human bladder cancer marker AG-CD71 and antibody ABC71 and application thereof
US11912749B2 (en) 2015-07-01 2024-02-27 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
US11912748B2 (en) 2015-07-01 2024-02-27 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6236864B2 (en) * 2012-06-11 2017-11-29 東ソー株式会社 Method for detecting cancer and antibody recognizing pancreatic ribonuclease 1
CN103018461B (en) * 2012-12-13 2015-03-18 中国科学院生物物理研究所 Kit for early diagnosis of bladder cancer and preparation method of kit
CN103033625B (en) * 2012-12-19 2014-12-24 中国科学院生物物理研究所 Human bladder cancer cellular chemiluminescent detection kit and preparation method thereof
CN104142401B (en) * 2014-07-25 2016-04-20 北京普恩光德生物科技开发有限公司 Tumor of bladder related antigen detection kit
WO2016026143A1 (en) 2014-08-22 2016-02-25 Huiru Wang Saccharide-based biomarkers and therapeutics
CN104267185B (en) * 2014-09-12 2016-05-18 范飞舟 Detect the kit of tumour and the material of special identification 2-Acetamido-2-deoxy-D-glucose thereof
CN104198707B (en) * 2014-09-12 2016-09-14 范飞舟 2-Acetamido-2-deoxy-D-glucose application in the test kit of preparation detection tumor
CN104761634B (en) * 2015-03-27 2018-05-18 李翀 A kind of pulmonary cancer diagnosis marker, antibody and its application
CN105859872A (en) * 2016-03-08 2016-08-17 李翀 Marker of human endometrial cancer, antibody and application of antibody
CN108676884A (en) * 2018-06-04 2018-10-19 海口市人民医院(中南大学湘雅医学院附属海口医院) Tumor marker for predicting bladder cancer
CN111254182B (en) * 2020-03-30 2024-01-02 深圳市兰科植物保护研究中心 Dendrobium polysaccharide anticancer activity analysis method and application of dendrobium polysaccharide

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4281061A (en) * 1979-07-27 1981-07-28 Syva Company Double antibody for enhanced sensitivity in immunoassay
US4761406A (en) * 1985-06-06 1988-08-02 The Procter & Gamble Company Regimen for treating osteoporosis
US20080076137A1 (en) * 2006-07-26 2008-03-27 Nicolai Bovin Bioanalytic Systems and Methods
WO2010007213A1 (en) * 2008-07-18 2010-01-21 Suomen Punainen Risti, Veripalvelu A method of evaluating the integrity of the plasma membrane of cells by detecting glycans found only intracellularly

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2003265556A1 (en) * 2002-08-20 2004-03-11 Millenium Pharmaceuticals, Inc. Compositions, kits, and methods for identification, assessment, prevention, and therapy of cervical cancer
EP1752536A4 (en) * 2004-05-11 2008-04-16 Alphagen Co Ltd Polynucleotide causing rna interfere and method of regulating gene expression with the use of the same
NZ545243A (en) * 2006-02-10 2009-07-31 Pacific Edge Biotechnology Ltd Urine gene expression ratios for detection of cancer
CN101768214B (en) * 2008-10-06 2013-03-13 复旦大学 Human tumor marker-Tim17 polypeptide and application thereof
CA2757625C (en) 2009-04-11 2017-11-28 Biotix, Inc. Automated pipette tip loading devices and methods
CN101638429B (en) * 2009-08-28 2012-01-25 南方医科大学 Small molecular peptide of specificity recognition cell surface integrin alphal3beta1

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4281061A (en) * 1979-07-27 1981-07-28 Syva Company Double antibody for enhanced sensitivity in immunoassay
US4761406A (en) * 1985-06-06 1988-08-02 The Procter & Gamble Company Regimen for treating osteoporosis
US20080076137A1 (en) * 2006-07-26 2008-03-27 Nicolai Bovin Bioanalytic Systems and Methods
WO2010007213A1 (en) * 2008-07-18 2010-01-21 Suomen Punainen Risti, Veripalvelu A method of evaluating the integrity of the plasma membrane of cells by detecting glycans found only intracellularly

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
Galustian et al. L-selectin Interactions with Novel Mono- and Multisulfated Lewisx Sequences in Comparison with the Potent Ligand 3*-Sulfated Lewisa. J. Biol. Chem. 1999, 274:18213-18217. *
JAMASBI et al. A Monoclonal Antibody to a Carbohydrate Epitope Expressed on Glycolipid and on alpha3beta1 Integrin on Human Esophageal Carcinoma. HYBRIDOMA AND HYBRIDOMICS. 22(6):367-376, 2003. *
Lityñska et al. Differences of alpha3ß1 integrin glycans from different human bladder cell lines. Acta Biochimica Polonica 47(2):427-434, 2000. *
Litynska et al. Profiling of integrin alpha3 beta1 N-linked oligosaccharides using DNA-sequencer. International Symposium on Predictive Oncology and Intervention Strategies; Paris, France; February 9 - 12, 2002; in the section on Molecular Pathology. Page 1 *
Pochec´ et al. Characterization of the oligosaccharide component of alpha3ß1 integrin from human bladder carcinoma cell line T24 and its role in adhesion and migration. European Journal of Cell Biology 85 (2006) 47-57. *
Prokopishyn et al. Integrin alpha3ß1 Expressed by Human Colon Cancer Cells Is A Major Carrier of Oncodevelopmental Carbohydrate Epitopes. Journal of Cellular Biochemistry 72:189-209 (1999). *
TAKEUCHI et al. InterceIIUlar AdheSlon Induced by Anti-alpha3 Integrln (VLA-3) Antibodles. EXPERIMENTAL CELL RESEARCH 21 1, 133-141 (1994). *
Zhao et al. N-Acetylglucosaminyltransferase III Antagonizes the Effect of N-Acetylglucosaminyltransferase V on alpha3beta1 Integrin-mediated Cell Migration. The Journal of Biological Chemistry, 281, 32122-32130, 2006. *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11912749B2 (en) 2015-07-01 2024-02-27 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
US11912748B2 (en) 2015-07-01 2024-02-27 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
JP2019513410A (en) * 2016-02-19 2019-05-30 ビーアンドエイチ バイオテクノロジーズ,エルエルシーB&H Biotechnologies, Llc Anti-N-acetylglucosamine and N-acetylgalactosamine antibodies
US11578134B2 (en) * 2017-08-14 2023-02-14 Chong Li Human bladder cancer marker AG-CD71 and antibody ABC71 and application thereof
WO2020257296A1 (en) * 2019-06-21 2020-12-24 Board Of Regents, The University Of Texas System Targeting alpha3beta1 integrin for treatment of cancer and other diseases
CN114641310A (en) * 2019-06-21 2022-06-17 德克萨斯大学***董事会 Targeted alpha 3 beta 1 integrins for the treatment of cancer and other diseases

Also Published As

Publication number Publication date
CN102372773B (en) 2013-06-05
US9073995B2 (en) 2015-07-07
CN102372773A (en) 2012-03-14
WO2012019437A1 (en) 2012-02-16

Similar Documents

Publication Publication Date Title
US9073995B2 (en) Bladder cancer tumor marker, antibody and use thereof
Song et al. Pharmacologic suppression of B7-H4 glycosylation restores antitumor immunity in immune-cold breast cancers
US7276372B2 (en) Antibodies against GPR64 and uses thereof
EP2799452B1 (en) Anti-human trop-2 antibody exhibiting antitumor activity in vivo
US9062100B2 (en) Anti-human TROP-2 antibody having anti-tumor activity in vivo
US9316654B2 (en) TAZ/WWTR1 for diagnosis and treatment of cancer
US20180362626A1 (en) csPCNA Isoform Antibodies And Uses Thereof
Min et al. L1 cell adhesion molecule is a novel therapeutic target in intrahepatic cholangiocarcinoma
US20010009665A1 (en) Individualized anti-cancer antibodies
EP2682475A1 (en) Antibody and antigen recognizing tumor-initiating cells and use thereof
JP2005508839A (en) Antibodies against peptides and MUC1 protein
CN101778867A (en) Compositions and methods for treating pancreatic tumors
Balmana et al. Analysis of sialyl-Lewis x on MUC5AC and MUC1 mucins in pancreatic cancer tissues
EP3750913B1 (en) Igg epitope and application thereof as target
Kwon et al. Monoclonal antibody targeting of the cell surface molecule TM4SF5 inhibits the growth of hepatocellular carcinoma
An et al. A novel anti-sTn monoclonal antibody 3P9 inhibits human xenografted colorectal carcinomas
US10444237B2 (en) Anti-pancreatic cancer monoclonal antibody and use thereof
US20150132291A1 (en) Methods for treatment of gastric cancer
EP1360208A1 (en) Induvidualized anti-cancer antibodies
WO2007122820A1 (en) Diagnostic agent and therapeutic agent for pancreatic cancer
Padhiar et al. Upregulated β1-6 branch N-glycan marks early gliomagenesis but exhibited biphasic expression in the progression of astrocytic glioma
US20220048980A1 (en) csPCNA Isoform Antibodies And Uses Thereof
Guan et al. Loss of caveolin-1 in cancer associated fibroblasts promotes hepatocellular carcinoma development
JP6182149B2 (en) Use of monoclonal antibody against EBAG9, hybridoma, and monoclonal antibody against EBAG9
WO2016155420A1 (en) Use of antibody specifically targeting wnt16b in preparation of anti-tumor medicine

Legal Events

Date Code Title Description
AS Assignment

Owner name: INSTITUTE OF BIOPHYSICS, CHINESE ACADEMY OF SCIENC

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LI, CHONG;FAN, ZUSEN;ZHANG, HONGLIAN;AND OTHERS;REEL/FRAME:029879/0222

Effective date: 20130218

STCF Information on status: patent grant

Free format text: PATENTED CASE

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YR, SMALL ENTITY (ORIGINAL EVENT CODE: M2551); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

Year of fee payment: 4

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YR, SMALL ENTITY (ORIGINAL EVENT CODE: M2552); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

Year of fee payment: 8